From 6f25367f24551a78bc3fe713a208eace48aaf82a Mon Sep 17 00:00:00 2001 From: Caroline Eastwood Date: Mon, 16 Dec 2024 11:44:36 +0000 Subject: [PATCH] Fixes #2851 Fixes #2851 - Edited the file: src/patterns/dosdp-patterns/ExtendedDescription.yaml to fix the typo. - Reran the command: ./run.sh make ../patterns/definitions.owl to update definition.owl. --- src/patterns/definitions.owl | 367 +++++++++--------- .../dosdp-patterns/ExtendedDescription.yaml | 2 +- 2 files changed, 174 insertions(+), 195 deletions(-) diff --git a/src/patterns/definitions.owl b/src/patterns/definitions.owl index 667aebe51..1b4b15e6e 100644 --- a/src/patterns/definitions.owl +++ b/src/patterns/definitions.owl @@ -7,8 +7,8 @@ Prefix(rdfs:=) Ontology( - -Annotation(owl:versionInfo "2024-12-06") + +Annotation(owl:versionInfo "2024-12-16") Declaration(Class()) Declaration(Class()) @@ -55,7 +55,6 @@ Declaration(Class()) Declaration(ObjectProperty()) Declaration(ObjectProperty()) Declaration(AnnotationProperty()) -Declaration(AnnotationProperty()) Declaration(AnnotationProperty()) Declaration(AnnotationProperty()) Declaration(AnnotationProperty()) @@ -68,7 +67,7 @@ Declaration(AnnotationProperty( () -AnnotationAssertion(Annotation( "DOI:10.1016/j.coi.2005.11.008") Annotation( "DOI:10.1038/nri3087") Annotation( "DOI:10.3389/fimmu.2019.01893") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.coi.2005.11.008") Annotation( "DOI:10.1038/nri3087") Annotation( "DOI:10.3389/fimmu.2019.01893") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Mononuclear phagocytes are immune cells that form a critical part of the body's innate immune system, the body's first line of defense against infections. Mononuclear phagocytes are characterized by the presence of a single, large nucleus. The ‘Mononuclear Phagocyte System’ nomenclature was introduced to distinguish mononuclear monocytes and macrophages from other cells with multiple nuclei. However, the cells within the mononuclear phagocyte system represent a highly heterogeneous group, all of which are able to perform highly efficient phagocytosis - engulfing and digesting microbes and cellular debris to fight infections and to maintain normal tissue homeostasis. In addition to phagocytosis, mononuclear phagocytes also secrete chemical compounds to recruit other immune cells to a site of infection. These cells produce different types of signaling molecules, including cytokines, chemokines, and reactive oxygen species, triggering a cascade of responses to ensure rapid and efficient containment and neutralization of invading pathogens. These cells are also involved in antigen presentation, a process essential for stimulating an adaptive immune response. They can process and present antigens to T cells, thereby linking the innate and adaptive immune responses. @@ -77,7 +76,7 @@ AnnotationAssertion( () -AnnotationAssertion(Annotation( "DOI:10.1002/glia.24343") Annotation( "DOI:10.1038/nn1988") Annotation( "DOI:10.1101/cshperspect.a020602") Annotation( "DOI:10.1126/science.aat0473") Annotation( "DOI:10.3389/fncel.2017.00024") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/glia.24343") Annotation( "DOI:10.1038/nn1988") Annotation( "DOI:10.1101/cshperspect.a020602") Annotation( "DOI:10.1126/science.aat0473") Annotation( "DOI:10.3389/fncel.2017.00024") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Glial cells, also known as neuroglia or simply glia, are non-neuronal cells in the central and peripheral nervous systems that provide support and protection for neurons. They constitute approximately half of the total cells in the human brain and more than half in other parts of the nervous system. Glial cells perform several key functions including, but not limited to, maintaining homeostasis, forming the myelin sheath around the neuron axons, and providing support and nutrition to neurons. These different functions are performed by various glial cell types, including astrocytes, oligodendrocytes, microglia and other specialized types. Astrocytes, for example, are the most abundant glial cells and provide metabolic and nutrient support to neurons, help regulate the extracellular ion and neurotransmitter levels, and play a role in the formation and maintenance of the blood-brain barrier, contributing to the overall homeostasis and functioning of the nervous system. Additionally, astrocytes are involved in synaptic communication and participate in processes such as synaptogenesis and synaptic pruning. @@ -88,7 +87,7 @@ AnnotationAssertion( () -AnnotationAssertion(Annotation( "DOI:10.1016/0166-4328(96)00075-7") Annotation( "DOI:10.1016/j.cell.2017.05.034") Annotation( "DOI:10.1038/s41574-019-0168-8") Annotation( "DOI:10.1073/pnas.1804938115") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/0166-4328(96)00075-7") Annotation( "DOI:10.1016/j.cell.2017.05.034") Annotation( "DOI:10.1038/s41574-019-0168-8") Annotation( "DOI:10.1073/pnas.1804938115") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Type EC enteroendocrine cells, also known as enterochromaffin cells, are a vital hormone-secreting cell type found in the gastrointestinal tract. These cells are named after their location in the intestines (“entero”) and because they are stainable by chromium salts (“chromaffin”). The primary function of type EC enteroendocrine cells is to act as chemosensors and lies in their capacity to produce and secrete serotonin, also known as 5-hydroxytryptamine (5-HT). Serotonin is a neurotransmitter that plays a significant role in modulating motility, secretion, vasodilation, perception of pain, and appetite in the gastrointestinal system. However, its function is not limited to the gastrointestinal tract; once secreted, serotonin is distributed via the bloodstream and contributes to regulating mood, appetite, and sleep in the brain. @@ -97,7 +96,7 @@ AnnotationAssertion( () -AnnotationAssertion(Annotation( "DOI:10.1007/s00424-017-1965-3") Annotation( "DOI:10.1016/j.cellimm.2018.01.005") Annotation( "DOI:10.1038/nri3600") Annotation( "DOI:10.14348/molcells.2021.0058") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00424-017-1965-3") Annotation( "DOI:10.1016/j.cellimm.2018.01.005") Annotation( "DOI:10.1038/nri3600") Annotation( "DOI:10.14348/molcells.2021.0058") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Alveolar macrophages are unique tissue-resident macrophages found in the lungs, specifically in the air sacs or alveoli where gas exchange occurs. They are characterized by specific surface markers including: F4/80-positive, CD11b-/low, CD11c-positive, CD68-positive, sialoadhesin-positive, dectin-1-positive, MR-positive, CX3CR1-negative. These specialized immune cells form a crucial part of the body's defense mechanism, playing important roles in pulmonary health and homeostasis. They are the first line of defense in the pulmonary immune response, acting as scavengers that patrol the alveoli and engulf foreign particles like bacteria, dust, and other debris that enter the lungs through inhalation. The primary function of these alveolar macrophages is phagocytosis, whereby they consume and digest foreign substances, dead cells, and other particulates. In the lung, alveolar macrophages are also responsible for clearing surfactant. Additionally, alveolar macrophages also play an integral role in initiating the immune response, as they secrete several pro-inflammatory cytokines and chemokines that recruit other immune cells to the site of infection or inflammation. @@ -106,7 +105,7 @@ AnnotationAssertion( () -AnnotationAssertion(Annotation( "DOI:10.1016/j.it.2022.10.010") Annotation( "DOI:10.1038/s41577-020-0281-x") Annotation( "DOI:10.3389/fimmu.2019.00036") Annotation( "DOI:10.3389/fphys.2020.00509/full") Annotation( "DOI:10.3892/mco.2017.1356") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.it.2022.10.010") Annotation( "DOI:10.1038/s41577-020-0281-x") Annotation( "DOI:10.3389/fimmu.2019.00036") Annotation( "DOI:10.3389/fphys.2020.00509/full") Annotation( "DOI:10.3892/mco.2017.1356") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial cells of lymphatic vessels are specialized type of cells that form the interior lining of the lymphatic system, which primarily comprises the lymph vessels and nodes. These cells play a critical role in creating a barrier between the lymphatic system and the surrounding tissues. They are structurally different from the endothelial cells of the blood vessels due to the presence of anchoring filaments and lack of a continuous basement membrane, and have a unique phenotype marked by the expression of various cell-specific markers such as Prox-1, VEGFR-3, and LYVE-1. A significant function of these endothelial cells is to maintain the fluid balance within the body. They facilitate the uptake of excess interstitial fluid that collects in the body tissues, and ensure its transport back into the bloodstream via the lymphatic vessels. These cells also enable the absorption of fats and fat-soluble vitamins from the digestive system, and their subsequent transport in the form of chyle, a milky fluid, to the blood. @@ -116,7 +115,6 @@ AnnotationAssertion( (cycling B cell) AnnotationAssertion(Annotation( "PMID:31348891") "A(n) B cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:31348891") "proliferating B cell") AnnotationAssertion(rdfs:label "cycling B cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -124,7 +122,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling T cell) AnnotationAssertion(Annotation( "PMID:37291214") "A(n) T cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:37291214") "proliferating T cell") AnnotationAssertion(rdfs:label "cycling T cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -132,7 +129,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling dendritic cell) AnnotationAssertion(Annotation( "PMID:32066951") "A(n) dendritic cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:32066951") "proliferating dendritic cell") AnnotationAssertion(rdfs:label "cycling dendritic cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -140,7 +136,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling natural killer cell) AnnotationAssertion(Annotation( "PMID:34062119") "A(n) natural killer cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34062119") "proliferating natural killer cell") AnnotationAssertion(rdfs:label "cycling natural killer cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -148,7 +143,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling gamma-delta T cell) AnnotationAssertion(Annotation( "PMID:32066951") "A(n) gamma-delta T cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:32066951") "proliferating gamma-delta T cell") AnnotationAssertion(rdfs:label "cycling gamma-delta T cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -156,7 +150,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling monocyte) AnnotationAssertion(Annotation( "PMID:31348891") "A(n) monocyte that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:31348891") "proliferating monocyte") AnnotationAssertion(rdfs:label "cycling monocyte") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -164,7 +157,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling CD8-positive, alpha-beta T cell) AnnotationAssertion(Annotation( "PMID:34062119") "A(n) CD8-positive, alpha-beta T cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34062119") "proliferating CD8-positive, alpha-beta T cell") AnnotationAssertion(rdfs:label "cycling CD8-positive, alpha-beta T cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -172,7 +164,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling CD4-positive, alpha-beta T cell) AnnotationAssertion(Annotation( "PMID:34062119") "A(n) CD4-positive, alpha-beta T cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34062119") "proliferating CD4-positive, alpha-beta T cell") AnnotationAssertion(rdfs:label "cycling CD4-positive, alpha-beta T cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -180,7 +171,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling macrophage) AnnotationAssertion(Annotation( "PMID:33208946") "A(n) macrophage that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:33208946") "proliferating macrophage") AnnotationAssertion(rdfs:label "cycling macrophage") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -188,7 +178,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling alveolar macrophage) AnnotationAssertion(Annotation( "PMID:37291214") "A(n) alveolar macrophage that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:37291214") "proliferating alveolar macrophage") AnnotationAssertion(rdfs:label "cycling alveolar macrophage") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -196,7 +185,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling mononuclear phagocyte) AnnotationAssertion(Annotation( "PMID:37468583") "A(n) mononuclear phagocyte that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:37468583") "proliferating mononuclear phagocyte") AnnotationAssertion(rdfs:label "cycling mononuclear phagocyte") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -204,7 +192,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling endothelial cell of lymphatic vessel) AnnotationAssertion(Annotation( "PMID:37291214") "A(n) endothelial cell of lymphatic vessel that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:37291214") "proliferating endothelial cell of lymphatic vessel") AnnotationAssertion(rdfs:label "cycling endothelial cell of lymphatic vessel") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -212,7 +199,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling pulmonary alveolar type 2 cell) AnnotationAssertion(Annotation( "PMID:37291214") "A(n) pulmonary alveolar type 2 cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:37291214") "proliferating pulmonary alveolar type 2 cell") AnnotationAssertion(rdfs:label "cycling pulmonary alveolar type 2 cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -220,7 +206,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling myeloid cell) AnnotationAssertion(Annotation( "PMID:38301653") "A(n) myeloid cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:38301653") "proliferating myeloid cell") AnnotationAssertion(rdfs:label "cycling myeloid cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -228,7 +213,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling basal cell) AnnotationAssertion(Annotation( "PMID:33208946") "A(n) basal cell that is cycling.") -AnnotationAssertion( "2024-07-02T09:14:23Z") AnnotationAssertion(Annotation( "PMID:33208946") "proliferating basal cell") AnnotationAssertion(rdfs:label "cycling basal cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -236,7 +220,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling stromal cell) AnnotationAssertion(Annotation( "PMID:34497389") "A(n) stromal cell that is cycling.") -AnnotationAssertion( "2024-07-19T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34497389") "proliferating stromal cell") AnnotationAssertion(rdfs:label "cycling stromal cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -244,7 +227,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling glial cell) AnnotationAssertion(Annotation( "PMID:34497389") "A(n) glial cell that is cycling.") -AnnotationAssertion( "2024-07-19T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34497389") "proliferating glial cell") AnnotationAssertion(rdfs:label "cycling glial cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -252,7 +234,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling plasma cell) AnnotationAssertion(Annotation( "PMID:34497389") "A(n) plasma cell that is cycling.") -AnnotationAssertion( "2024-07-19T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34497389") "proliferating plasma cell") AnnotationAssertion(rdfs:label "cycling plasma cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -260,7 +241,6 @@ EquivalentClasses( ObjectIntersection # Class: (cycling type EC enteroendocrine cell) AnnotationAssertion(Annotation( "PMID:34497389") "A(n) type EC enteroendocrine cell that is cycling.") -AnnotationAssertion( "2024-07-19T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34497389") "proliferating type EC enteroendocrine cell") AnnotationAssertion(rdfs:label "cycling type EC enteroendocrine cell") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) @@ -268,48 +248,47 @@ EquivalentClasses( ObjectIntersection # Class: (cycling neuroblast (sensu Vertebrata)) AnnotationAssertion(Annotation( "PMID:34497389") "A(n) neuroblast (sensu Vertebrata) that is cycling.") -AnnotationAssertion( "2024-07-19T09:14:23Z") AnnotationAssertion(Annotation( "PMID:34497389") "proliferating neuroblast (sensu Vertebrata)") AnnotationAssertion(rdfs:label "cycling neuroblast (sensu Vertebrata)") EquivalentClasses( ObjectIntersectionOf( ObjectSomeValuesFrom( ) ObjectSomeValuesFrom( ))) -AnnotationAssertion(Annotation( "DOI:10.1016/S0092-8674(00)81834-6") Annotation( "DOI:10.1016/S1534-5807(02)00173-9") Annotation( "DOI:10.1242/dev.202046") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553142/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S0092-8674(00)81834-6") Annotation( "DOI:10.1016/S1534-5807(02)00173-9") Annotation( "DOI:10.1242/dev.202046") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553142/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Male germ cells are specialized cells that give rise to the male gametes and are therefore crucial for sexual reproduction in males. Through a complex process called spermatogenesis in the testes male germ cells undergo several stages of development, each with distinctive morphological and physiological characteristics, eventually differentiating into mature spermatozoa (sperm). Male germ cells are responsible for transferring genetic material from male to offspring as part of sexual reproduction. In the initial stage of their life cycle, they exist as spermatogonial stem cells, which multiply by mitotic division to self-renew and to produce daughter cells. The daughter cells then undergo meiotic division, a process that reduces the number of chromosomes in each cell by half, from diploid to haploid – the formation of spermatids. This is critical for maintaining genetic stability from generation to generation, as it ensures that when an egg and sperm cell unite during fertilization, the resulting offspring will have the correct number of chromosomes. The final phase of spermatogenesis is called spermiogenesis, during which spermatozoa - mature and motile sperm cells – are produced from round spermatids. The matured male germ cells or sperm cells are highly specialized, having a streamlined shape designed for efficient movement towards the female egg. A sperm cell is divided into three main parts, namely the head, midpiece, and tail. The head contains the genetic material, the midpiece contains mitochondria that provide energy, and the tail, termed as the flagellum, propels the sperm cell forward. Each of these parts play significant roles in fertilization.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000015") -AnnotationAssertion(Annotation( "DOI:10.1080/19420889.2015.1017156") Annotation( "DOI:10.1152/physrev.00009.2020") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK26914/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1080/19420889.2015.1017156") Annotation( "DOI:10.1152/physrev.00009.2020") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK26914/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Sperm cells, also known as spermatozoa or male gametes, are highly specialized cells that are pivotal for sexual reproduction in animals. Produced in the male reproductive organs, specifically the testes, sperm cells play an important role in the process of fertilization, where they fuse with a female gamete or egg cell (ovum) to form a zygote, which marks the beginning of a new organism. Sperm cells hold several unique traits that set them apart from other body cells. Structurally, a sperm cell possesses two main structures: the head, containing the nucleus carrying the paternal genetic information, and the flagellum, which generates energy and grants the sperm cell mobility The process of fertilization occurs when one sperm cell successfully penetrates the egg, delivering its genetic information, and ultimately leading to the formation of a new individual combining both paternal and maternal genetic materials. Notably, while hundreds of millions of sperm cells may be released during ejaculation, only one has the opportunity to successfully fertilize the egg, underscoring the highly competitive nature of this biological process.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000019") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c120026") Annotation( "DOI:10.1007/978-1-4939-1311-4_10") Annotation( "DOI:10.1016/j.immuni.2022.08.002") Annotation( "DOI:10.1038/nri.2017.11") Annotation( "DOI:10.1111/j.1478-3231.2006.01342.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c120026") Annotation( "DOI:10.1007/978-1-4939-1311-4_10") Annotation( "DOI:10.1016/j.immuni.2022.08.002") Annotation( "DOI:10.1038/nri.2017.11") Annotation( "DOI:10.1111/j.1478-3231.2006.01342.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kupffer cells are tissue-resident macrophages located in the liver. They are an integral part of the mononuclear phagocyte system and are responsible for the phagocytosis of dead or dying cells, microbes, and other foreign substances. Kupffer cells account for approximately 80-90% of tissue-resident macrophages in the body. They are found within the lumen of liver sinusoids and interface with microbial populations and products. The primary role of Kupffer cells is to maintain homeostasis in the liver. They continuously filter and cleanse the blood that flows through the liver, removing pathogens, endotoxins, particulate matter, aged and dysfunctional red blood cells, and miscellaneous waste products. These cells also play an important role in maintaining iron homeostasis. During the process of phagocytosis, Kupffer cells recycle iron from degraded red blood cells, which is then utilized in the formation of new erythrocytes. Kupffer cells also play key roles in various immune responses and inflammatory processes. They secrete various types of cytokines and chemokines, which contribute to the activation and mobilization of other immune cells. They can also produce reactive oxygen species and nitric oxide, which have microbicidal actions. Although their activity is essential for host defense, excessive or prolonged activation of Kupffer cells may contribute to hepatic injury, inflammation, and fibrosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000091") -AnnotationAssertion(Annotation( "DOI:10.3389/fmed.2017.00234") Annotation( "DOI:10.37175/stemedicine.v1i4.57") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/osteoclast") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.3389/fmed.2017.00234") Annotation( "DOI:10.37175/stemedicine.v1i4.57") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/osteoclast") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Osteoclasts, derived from the monocyte/macrophage lineage, play a crucial role in bone remodeling through bone resorption. They are distinguished from other cells by their large size and their unique ability to secrete enzymes and acids that dissolve the calcium phosphate in bone. The formation of osteoclasts is influenced by two primary factors: macrophage colony-stimulating factor (M-CSF) and the ligand-activated receptor (RANKL) of nuclear factor kappa B (NF-κB). Osteoclasts also collaborate with osteoblasts, responsible for new bone formation, to maintain bone health and adapt bone structure. Their coordinated actions allow continuous remodeling while preserving overall bone strength and integrity. Imbalances in osteoclast activity can lead to pathological conditions. For example, excessive activity may contribute to diseases like osteoporosis, periprosthetic osteolysis, bone tumors, and Paget's disease. Other the other hand, osteoclast deficiency ca result in conditions like osteopetrosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000092") -AnnotationAssertion(Annotation( "DOI:10.1038/srep32007") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK539836/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK563130") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/srep32007") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK539836/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK563130") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Surface ectoderm cells give rise to external structures such as the epidermis, hair, nails, and sweat glands. They are critical during the early stages of embryonic development, where they delineate and create the organism's exterior body plan. Beyond forming the organism's physical exterior, surface ectodermal cells have other significant roles. For instance, these cells lead to the development of the anterior pituitary gland and the enamel of the teeth. They are also integral to the formation of the sensory organs, including the eyes and the ears. In the eyes, they form the cornea and lens epithelium. In the ears, they form the external ear canal and certain parts of the inner ear. Therefore, these cells are instrumental in the development of various sensory systems in an organism. Certain abnormalities or mutations in surface ectodermal cells could lead to medical conditions known as ectodermal dysplasias. These are a group of inherited disorders affecting the development or function of teeth, hair, nails, and sweat glands. Therefore, understanding the formation and function of surface ectodermal cells is also important in identifying and addressing certain genetic disorders and conditions. In summary, surface ectodermal cells are essential in embryonic development, contributing to the creation of the external physical appearance and the sensory organs of an organism.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000114") -AnnotationAssertion(Annotation( "DOI:10.1007/s12311-018-0985-7") Annotation( "DOI:10.1038/nrn3886") Annotation( "DOI:10.7554/eLife.63668") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK545154/") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/purkinje-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s12311-018-0985-7") Annotation( "DOI:10.1038/nrn3886") Annotation( "DOI:10.7554/eLife.63668") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK545154/") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/purkinje-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Purkinje cells, named after the Czech anatomist Jan Evangelista Purkyně who discovered them, are unique inhibitory neurons in the cerebellar cortex. They are a critical part of the vertebrate nervous system as they provide the only signal output from the cortex to the cerebellar nuclei. They are one of the few types of neurons that are large enough to be seen with the naked eye. The most distinct hallmark of Purkinje cells is their elaborate dendritic arbor, which forms a broad and intricately branching structure resembling a tree. These numerous branches each receive excitatory synaptic inputs from more than 100,000 parallel fibers; in addition, a single climbing fiber makes hundreds of synapses to the soma and proximal dendrites. A single long axon forms an inhibitory projection to the cerebellar nuclei. Purkinje cells play key roles in the coordination of fine, voluntary motor movements and balance. As the sole output of all motor coordination in the cerebellar cortex, they serve as a central relay in the cerebro-cerebellar loop. Each Purkinje cell receives two types of synaptic input: one from parallel fibers (which are axons of granule cells), and the other from climbing fibers (originating from the inferior olivary nucleus). The Purkinje cells process and integrate these diverse kinds of input signals to generate output that controls timing and coordination of movements. Purkinje neurons show considerable synaptic plasticity. Throughout life, these cells continue to undergo long-term potentiation and depression at parallel fiber synapses, which cause long-lasting increase or decrease, respectively, of synaptic transmission and have been proposed as mechanisms for motor learning. Purkinje cells are also known to be implicated in a variety of diseases. Their progressive loss is a prime feature in certain types of ataxia, a collective term used to describe conditions characterized by loss of muscular control and coordination. Furthermore, a significant reduction in the density of Purkinje cells has been reported in conditions such as autism and Huntington’s disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000121") -AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2019.08.053") Annotation( "DOI:10.1016/j.conb.2022.102674") Annotation( "DOI:10.1016/j.tins.2021.11.001") Annotation( "DOI:10.1038/nri3086") Annotation( "DOI:10.3389/fnins.2021.742065") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2019.08.053") Annotation( "DOI:10.1016/j.conb.2022.102674") Annotation( "DOI:10.1016/j.tins.2021.11.001") Annotation( "DOI:10.1038/nri3086") Annotation( "DOI:10.3389/fnins.2021.742065") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Microglial cells, sometimes referred to as microglia, are a type of glial cell that primarily exist within the central nervous system (CNS), notably in the brain and the spinal cord. Classified among the resident immune cells, microglial cells represent about 10% of all cells found within the CNS. These cells are derived from progenitor cells in the yolk sac, which differentiates them from other types of glial cells (such as astrocytes and oligodendrocytes) that are derived from neuroectodermal cell lineages. The primary role of microglial cells is to act as the first and main active form of immune defense in the CNS. They express a vast repertoire of pattern recognition receptors, which allow them to sense and eliminate microbes invading the CNS parenchyma. They represent one of the macrophage populations of the CNS and are responsible for phagocytosis (engulfing and destroying cellular waste or pathogens) in the neural environment. @@ -317,156 +296,156 @@ Microglial cells are particularly responsive to pathogens and injuries and chang Beyond their macrophagic activity, they also perform synaptic pruning during brain development, eliminate unnecessarily produced neurons, and facilitate tissue regeneration and repair. they play integral roles in regulating neural development and supporting cell survival and are important for maintaining tissue homeostasis. While their protective role generally benefits the brain, their over-activation can occasionally lead to neuro-inflammatory diseases, underlining the importance of balanced microglial cell functions. They have been extensively studied for their association with neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000129") -AnnotationAssertion(Annotation( "DOI:10.1038/nri2171") Annotation( "DOI:10.1111/bph.14527") Annotation( "DOI:10.3390/ijms23073698") Annotation( "DOI:10.3748/wjg.v17.i5.578") Annotation( "DOI:full/10.15252/emmm.202114121") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nri2171") Annotation( "DOI:10.1111/bph.14527") Annotation( "DOI:10.3390/ijms23073698") Annotation( "DOI:10.3748/wjg.v17.i5.578") Annotation( "DOI:full/10.15252/emmm.202114121") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Gut endothelial cells are a crucial component of the endothelium, a thin layer of single cells that line the interior surface of blood vessels and lymphatic vessels, including those in the digestive system or \"gut\". These cells plays an integral role in various physiological and metabolic functions, actively participating in nutrient absorption, host defense, and vascular homeostasis in the gut. The primary function of gut endothelial cells lies in their ability to control the passage of materials and the transit of white blood cells into and out of the bloodstream. They achieve this through the formation of a semi-permeable barrier, in which permeability is regulated by tight junctions; complex structures that bring the cells together, sealing the space between them. This ensures a controlled, selective passage of nutrients, ions, and water from the bloodstream into the gut and vice versa, helping maintain homeostasis and overall health. Gut endothelial cells also play a significant role in angiogenesis and serve as a critical regulator of the gut immune responses playing a modulative role in gut immune homeostasis and inflammatory responses. In addition, they generate nitric oxide, a potent vasodilator, which helps in maintaining vascular tonus, preventing platelet and leukocyte adhesion, and decreasing smooth muscle proliferation. These multi-faceted roles make gut endothelial cells indispensable for the normal functioning of the gut and overall health.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000131") -AnnotationAssertion(Annotation( "DOI:10.1002/med.21754") Annotation( "DOI:10.1038/nrc.2016.37") Annotation( "DOI:10.1111/j.1751-1097.2007.00226.x") Annotation( "DOI:10.3390/ijms21249769") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/melanocyte") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/med.21754") Annotation( "DOI:10.1038/nrc.2016.37") Annotation( "DOI:10.1111/j.1751-1097.2007.00226.x") Annotation( "DOI:10.3390/ijms21249769") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/melanocyte") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Melanocytes are specialized cells prevalently found in the skin, but also present in the hair follicles, eyes, inner ear, bones, heart, and brain. Their primary function is the synthesis of melanin, a pigment responsible for coloration of skin, hair, and eyes. The presence and distribution of this pigment play a crucial role in the body's defense mechanism against harmful ultraviolet radiation. In addition to melanin production, melanocytes also engage in a variety of other biological functions. They play a fundamental role in the immune response, owing to their ability to present antigens to T cells. Furthermore, they contribute to the maintenance of the skin's homeostasis by regulating processes such as extracellular matrix remodeling and cytokine production. A balance in melanocyte function is pivotal, as dysfunctions often result in dermatological disorders. For instance, the loss of melanocytes or reduction in their function may lead to hypopigmentation conditions such as vitiligo, whereas an abnormal increase could result in hyperpigmentation disorders or melanoma, a dangerous type of skin cancer.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000148") -AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1038/s41575-022-00675-x") Annotation( "DOI:10.1165/ajrcmb.25.5.f218") Annotation( "DOI:10.1242/bio.20121701") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553208/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1038/s41575-022-00675-x") Annotation( "DOI:10.1165/ajrcmb.25.5.f218") Annotation( "DOI:10.1242/bio.20121701") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553208/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Goblet cells are specialized, simple columnar, secretory epithelial cells that are mostly found in the respiratory and intestinal tracts. The term \"goblet\" refers to their shape, which resembles a flask or goblet, specially at their apical end which is swollen due to the accumulation of secretory granules. The primary function of goblet cells is to to protect and lubricate the underlying tissues by secreting large quantities of mucin, a complex glycoprotein, which forms mucus when hydrated. In the respiratory tract, the mucus secreted by these cells traps dust, bacteria, viruses, and other potentially harmful particles in the inhaled air, preventing them from reaching the delicate tissues of the lungs. In the intestines, the goblet cells secrete mucus that acts as a protective barrier shielding the intestinal epithelium from dietary antigens, pathogens and prevents the intestinal epithelium from being eroded by the actions of the digestive enzymes and the abrasive action of passing food material. Goblet cells are capable of rapidly altering their secretory output in response to stimuli. For example, irritants like smoke or dust can trigger an increased rate of mucus production, as the body attempts to flush out the harmful particles. Conversely, in conditions such as chronic bronchitis and cystic fibrosis, overactive goblet cells can create a thick accumulation of mucus that obstructs the airways and fosters bacterial growth. Lastly, goblet cells are not static, rather, they undergo a dynamic process known as goblet cell metaplasia-differentiation, wherein non-goblet cells in response to chronic injury or inflammation, can differentiate into goblet cells leading to an accumulation of these cells in the tissue, known as goblet cell hyperplasia.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000160") -AnnotationAssertion(Annotation( "DOI:10.1038/s41574-019-0168-8") Annotation( "DOI:10.1111/j.1463-1326.2011.01438.x") Annotation( "DOI:10.1196/annals.1294.001") Annotation( "DOI:10.1210/endrev/bnaa018") Annotation( "DOI:10.7554/elife.78512") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41574-019-0168-8") Annotation( "DOI:10.1111/j.1463-1326.2011.01438.x") Annotation( "DOI:10.1196/annals.1294.001") Annotation( "DOI:10.1210/endrev/bnaa018") Annotation( "DOI:10.7554/elife.78512") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Enteroendocrine cells are a specialized subset of cells located within the epithelial lining of both the small and large intestines, as well as the stomach and pancreas. Constituting less than 1% of the total population of intestinal cells, they are differentiated from a common intestinal cell progenitor, thus sharing lineage with absorptive and goblet cells of the intestines. The distinct characteristic feature of an enteroendocrine cell is its inherent capacity to synthesize and secrete a plethora of gut hormones such as serotonin, somatostatin, neurotensin, cholecystokinin, secretin, gastric inhibitory peptide, motilin, and glucagon-like peptide-1. Integrated within this complex network of signaling agents, these substances control various aspects of the digestive system. Each enteroendocrine cell has its unique combination of hormones to release, determined by its position along the intestinal tract. Intriguingly, these hormones not only modulate local gut function, including motility, absorption and secretion, but also potentiate distant actions on other systems such as endocrine, nervous and immune and play a role in the feeling of satiety. The release of these hormones from enteroendocrine cells is a highly regulated and dynamic process. The cells are equipped with sensory receptors localized on its luminal side that respond to various stimuli, including changes in nutrient composition, chemical or mechanical changes in the gut lumen, or even signals arising from commensal microbiota. This sensory input stimulates a signaling cascade within the cell, culminating in the release of specific hormones into the interstitial fluid. These hormones then make their way into the bloodstream, acting on their respective target receptors to mediate their duties. The multifunctional characteristics of enteroendocrine cells make them crucial for maintaining gut homeostasis and the overall physiological well being of the body.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000164") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c190003") Annotation( "DOI:10.1016/j.biocel.2016.02.003") Annotation( "DOI:10.3389/fendo.2018.00711") Annotation( "DOI:10.3389/fimmu.2022.977175") Annotation( "DOI:10.3389/fimmu.2022.977175/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c190003") Annotation( "DOI:10.1016/j.biocel.2016.02.003") Annotation( "DOI:10.3389/fendo.2018.00711") Annotation( "DOI:10.3389/fimmu.2022.977175") Annotation( "DOI:10.3389/fimmu.2022.977175/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Chromaffin cells, also known as pheochromocytes, are neuroendocrine cells that are typically located in the adrenal medulla, the innermost part of the adrenal gland, which is situated on top of each kidney. Chromaffin cells are also found in small clusters, known as paraganglia, in various locations throughout the body, including the sympathetic nervous system. They derive their name from their ability to stain a brownish-black color upon exposure to chromic salts, a feature made possible due to their high content of granules rich in catecholamines and catecholamine-related neurotransmitters. The primary function of chromaffin cells is the synthesis and release of catecholamines, specifically epinephrine (adrenaline) and norepinephrine (noradrenaline). These neurotransmitters are vital stress hormones that, when released by the adrenal chromaffing cells into the bloodstream, prepare the body for the 'fight or flight' response. This response can enhance the body's performance in a dangerous situation by increasing heart rate, elevating blood sugar, and increasing blood flow to the muscles. The chromaffin cells in paraganglia are responsible for the local release of catecholamines and play a role in regulating blood pressure and other autonomic functions. In addition to their role in stress response, chromaffin cells also contribute to the body's immune response. They secrete several peptides including antimicrobial peptides, and the discovery of LPS and cytokine receptors on chromaffin cells suggests that the adrenal medulla may participate in some aspects of the immune response.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000166") -AnnotationAssertion(Annotation( "DOI:10.1016/j.coemr.2019.03.001") Annotation( "DOI:10.1093/biolre/ioy059") Annotation( "DOI:10.1210/clinem/dgaa603") Annotation( "DOI:10.3389/fendo.2014.00006") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556007/#:~:text=Leydig%20cells%20are%20the%20primary,secondary%20sexual%20characteristics%20and%20behaviors.") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.coemr.2019.03.001") Annotation( "DOI:10.1093/biolre/ioy059") Annotation( "DOI:10.1210/clinem/dgaa603") Annotation( "DOI:10.3389/fendo.2014.00006") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556007/#:~:text=Leydig%20cells%20are%20the%20primary,secondary%20sexual%20characteristics%20and%20behaviors.") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Leydig cells are a testosterone-secreting cell in the interstitial area in the testes of males. They are named after Franz Leydig, a German anatomist who discovered these cells in 1850. Uniquely situated within the soft connective tissue surrounding the seminiferous tubules, Leydig cells form an integral part of the male reproductive system. They are usually polygonal cells characterized by well developed smooth endoplasmic reticulum, high lipid content and a large round nucleus. They are found across mammalian species, including humans. The primary function of Leydig cells is the production of androgens, the male sex hormones, the most notable of which is testosterone. Leydig cells synthesize testosterone from cholesterol through a series of enzymatic reactions. The production and release of testosterone are mainly regulated by the luteinizing hormone (LH) released by the anterior pituitary gland. In response to LH, Leydig cells convert cholesterol into testosterone, which then plays a crucial role in the development and maintenance of primary and secondary male sexual characteristics. These include the formation and maturation of male reproductive organs, onset of spermatogenesis, and the presentation of male secondary sexual traits such as the deepening of the voice, growth of facial hair, development of muscles, and a broadening of shoulders. Apart from testosterone production, Leydig cells also secrete insulin-like factor 3 (INSL3) that is essential for testicular descent during embryonic development in males. Dysregulation or loss of Leydig cells can lead to numerous conditions like testosterone deficiency, infertility, and certain forms of testicular cancer. Although not typical, Leydig cells can regenerate if they are damaged, ensuring the continuous production of testosterone and maintaining male reproductive health. They provide an excellent model to study cell differentiation and hormone regulation, thereby enhancing our understanding of reproductive biology and associated disorders.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000178") -AnnotationAssertion(Annotation( "DOI:10.1016/j.biocel.2011.11.011") Annotation( "DOI:10.1055/s-2007-1007096") Annotation( "DOI:10.1083/jcb.201903090") Annotation( "DOI:10.1111/j.1439-0396.2007.00752.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.biocel.2011.11.011") Annotation( "DOI:10.1055/s-2007-1007096") Annotation( "DOI:10.1083/jcb.201903090") Annotation( "DOI:10.1111/j.1439-0396.2007.00752.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Hepatocytes are the major cell type constituting 70-80% of the liver's cytoplasmic mass, playing crucial roles in maintaining the body's metabolic homeostasis. Dimensions of mature hepatocytes typically range from 20 to 30 μm in humans, but size may vary depending on their location within the liver lobule. Hepatocytes are characterized by high biosynthetic, enzymatic, and endocytic activity. They contain abundant mitochondria, smooth and rough endoplasmic reticulum, peroxisomes, lysosomes, and a large nucleus that is often binucleate. The liver consists of three zones - the periportal Zone 1, midzone 2, and pericentral Zone 3 - which have differential nutrient and oxygen status, and damage susceptibility; hepatocytes in the different zones show signifcant functional heterogeneity ('hepatocyte functional zonation'). Hepatocytes are involved in a multitude of critical functions including the metabolism of lipids, carbohydrates, and proteins, the synthesis of serum proteins (e.g., albumin, transferrin, and lipoproteins), the detoxification and excretion of endogenous and exogenous substances, the storage of vitamins and minerals, and the production and secretion of bile. Heterocytes demonstrate a remarkable regenerative capacity, which enables the liver to recover from injury and loss of tissue mass. Notably, hepatocytes perform biotransformation with both phase I and phase II enzymes, which modify drugs, xenobiotics, and various substances for elimination from the body. Phase I enzymes, such as cytochrome P450, catalyze both oxidative and reductive reactions of many xenobiotics; many of the products of phase I enzymes are substrates for the phase II enzymes, which catalyze conjugation reactions. Alterations in hepatocyte function have significant implications for overall human health and disease. Certain conditions, such as hepatitis, cirrhosis, and liver cancer, can profoundly affect hepatocyte structure and function, thereby disrupting the liver's ability to perform its vital roles within the body. As the primary site for drug metabolism, changes in hepatocyte function can also impact the effectiveness and toxicity of pharmaceuticals. The regenerative ability of hepatocytes makes them valuable cells for liver regenerative medicine and bioartificial liver support systems, and their study has provided significant insights into liver biology and disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000182") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c160033") Annotation( "DOI:10.3390/biology10101056") Annotation( "https://www.nature.com/scitable/topicpage/the-sliding-filament-theory-of-muscle-contraction-14567666/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK537139") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK9961/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c160033") Annotation( "DOI:10.3390/biology10101056") Annotation( "https://www.nature.com/scitable/topicpage/the-sliding-filament-theory-of-muscle-contraction-14567666/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK537139") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK9961/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Skeletal muscle cells, or myocytes, are essential for the musculoskeletal system, featuring a striated appearance from tightly packed sarcomeres. These elongated, multi-nucleated cells convert ATP into mechanical energ and are key for voluntary movement and posture. They also are responsible for maintaining body temperature, storing nutrients, and stabilizing joints. Skeletal muscle cells organize themselves tightly into bundles to form muscle fibers. Embedded within each of these cells are thousands of myofibrils, which are made up of the contractile proteins, actin (the thin filament), and myosin (the thick filament). Contraction of a skeletal muscle cell occurs when these myofibrils shorten, a process driven by the sliding of actin and myosin filaments over each other in a mechanism famously known as the sliding filament theory. Additionally, skeletal muscle cells exhibit a high degree of plasticity, which allows for their adaptation in response to alterations in functional demands or damages. They have an innate regenerative capacity due to the presence of satellite cells—quiescent muscle stem cells that reside within the muscle fibers. Upon muscle injury, these satellite cells become activated, proliferate, and differentiate into new muscle cells, thereby contributing to the repair and growth of injured skeletal muscle. With this, skeletal muscle cells serve not only a pivotal biomechanical function but also possess a great capacity for self-healing and regeneration.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000188") -AnnotationAssertion(Annotation( "DOI:10.1093/ptj/81.11.1810") Annotation( "DOI:10.1186/s13578-015-0054-6") Annotation( "DOI:10.3389/fphys.2021.747214") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/slow-muscle-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1093/ptj/81.11.1810") Annotation( "DOI:10.1186/s13578-015-0054-6") Annotation( "DOI:10.3389/fphys.2021.747214") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/slow-muscle-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Slow muscle cells, as the name implies, are a type of muscle cell well known for their slow contractile speed. They are also referred to as type I, slow-twitch, or red muscles because of their high myoglobin content which gives them a dark, reddish appearance. These cells are predominantly found in the postural muscles of the body, such as those in the back and lower limbs. A defining characteristic of slow muscle cells is their significant resistance to fatigue. This is largely due to their efficient oxidative metabolism, which allows them to continuously contract over extended periods without succumbing to fatigue. This extraordinary endurance is facilitated by a high concentration of mitochondria, which enables the efficient use of oxygen for energy production, and capillaries, which supplies the necessary oxygen and nutrients. Additionally, these cells have an abundant supply of myoglobin, a protein that stores and transports oxygen within the muscle cell, further supporting their aerobic metabolism. The primary role of slow muscle cells in the human body is to provide sustained, low-intensity contractions over a prolonged period. They are responsible for maintaining posture and providing stability, rather than delivering short, powerful bursts of activity. They play a pivotal role in supporting various physical activities such as standing, walking, or any form of exercise that requires endurance over speed.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000189") -AnnotationAssertion(Annotation( "DOI:10.1152/japplphysiol.00636.2019") Annotation( "DOI:10.3389/fcell.2018.00125") Annotation( "DOI:10.4161/org.4.3.6312") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/fast-muscle-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/japplphysiol.00636.2019") Annotation( "DOI:10.3389/fcell.2018.00125") Annotation( "DOI:10.4161/org.4.3.6312") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/fast-muscle-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Fast muscle cells, also known as type II muscle fibers, are a particular type of muscle cell that specializes in conducting rapid, high-intensity contractions. These cells are primarily found within skeletal muscle groups that are directly involved in gross motor activities, such as running, jumping, or lifting heavy weights. The designation \"fast\" refers to their speed of contraction in response to neural stimuli, which is significantly quicker when compared to other muscle cell types such as slow-twitch or type I muscle fibers. The primary function of fast muscle cells revolves around their role in anaerobic metabolism, which provides the energy needed for short, forceful bursts of power. They contain a high concentration of glycolytic enzymes that facilitate this process, leading to a swift break down of glucose to generate ATP (adenosine triphosphate), the primary energy currency in biological systems. On the downside, the byproduct of this very rapid, anaerobic metabolic process is lactic acid, which can build up and cause muscle fatigue. Fast muscle cells can be further classified into two subtypes based on their metabolic characteristics: type IIa and type IIb/x. Type IIa cells, also known as fast oxidative-glycolytic fibers, possess a good oxygen supply and can function in both anaerobic and aerobic conditions, exhibiting moderate resistance to fatigue. On the other hand, type IIb/x cells, also known as fast glycolytic fibers, primarily rely on anaerobic metabolism and tire out a lot quicker. Both these subtypes can adapt to changing demands due to exercise and conditioning, highlighting the plasticity that is a key characteristic of these versatile muscle cells.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000190") -AnnotationAssertion(Annotation( "DOI:10.1093/ptj/81.11.1810") Annotation( "https://training.seer.cancer.gov/anatomy/muscular/types.html") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK10854/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK526125") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556137/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1093/ptj/81.11.1810") Annotation( "https://training.seer.cancer.gov/anatomy/muscular/types.html") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK10854/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK526125") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556137/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Smooth muscle cells are a specialized type of muscle cells that are primarily found within the walls of hollow organs such as the intestine, stomach, bladder, uterus, and blood vessels. These cells are an essential component of the autonomic nervous system controling involuntary movements within the body. Smooth muscle cells play a vital role in many physiological processes due to their unique functionality. They can contract and relax in a slow, sustained, and controlled manner to help modulate the size and shape of the lumens of the organs they surround. For example, in the digestive system, coordinated contraction and relaxation of smooth muscle cells propels food along the gastrointestinal tract - a process known as peristalsis. In the blood vessels, the smooth muscle cells, by contracting or relaxing, either constrict or dilate the vessels, which in turn, help regulate blood flow and pressure. On a cellular level, smooth muscle cells possess a single, centrally located nucleus and contain an arranged network of actin and myosin filaments, which are the proteins responsible for muscle contraction. Unlike their counterparts in skeletal or cardiac muscles, these cells lack specific contact points for these filaments, hence the lack of visible striations. Furthermore, the contraction of smooth muscle cells is regulated by hormones and neurotransmitters, such as acetylcholine and norepinephrine, adding an additional layer of complex regulation to these highly specialized cells.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000192") -AnnotationAssertion(Annotation( "DOI:10.1016/j.ceb.2019.07.012") Annotation( "DOI:10.5535/arm.2016.40.1.162") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/mesoderm") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.ceb.2019.07.012") Annotation( "DOI:10.5535/arm.2016.40.1.162") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/mesoderm") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Mesodermal cells are pluripotent and the most abundant in the human body. They contribute to the development of several structures such as the skeletal muscles, bones, heart and blood vessels, kidneys, gonads, connective tissues, and certain layers of the skin. Hence, the mesoderm is often regarded as an active and diverse layer due to its role in forming many bodily structures. In embryonic development, these cells are instrumental, contributing to the body's structures through a carefully coordinated sequence of morphogenetic movements. This process forms the complex multicellular architecture of tissues and organs from a simple ball of cells. Without the function of mesodermal cells, the body's integral structures and systems would cease to form correctly, leading to developmental disorders such as axial mesodermal dysplasia complex") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000222") -AnnotationAssertion(Annotation( "DOI:10.1038/s41586-020-2347-0") Annotation( "DOI:10.3389/fcell.2022.1040708") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK10008/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553142/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41586-020-2347-0") Annotation( "DOI:10.3389/fcell.2022.1040708") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK10008/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553142/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Gametes are reproductive cells essential for sexual reproduction in eukaryotic organisms, including humans, animals, and many plants. These cells are haploid, containing half the genome or only one set of chromosomes, which is a contrast to the diploid state (two sets of chromosomes) found in most cells in a multicellular organism. This reduction in genetic material is crucial since it allows for the re-establishment of the diploid number of chromosomes when two gametes merge to form a diploid zygote during fertilization. There are two major types of gametes in animals: sperm cells in males and egg (or ova) in females. These gametes contribute equally to the genetic makeup of the offspring but are morphologically and functionally distinct. Sperm cells (or spermatozoa) are characterized by their motility, small size, and large quantities; they are produced continuously in vast numbers through a process called spermatogenesis in the male gonads or testes. Female gametes are much larger, fewer in numbers, and are not motile; they undergo a development process known as oogenesis, which takes place within the ovaries. The egg also contributes most of the cytoplasm and organelles (including mitochondria) to the zygote that are required for initial cell division stages after fertilization. Gametes have a primary role in sexual reproduction and genetic variation, which aids in evolution. The formation of gametes involves a special type of cell division called meiosis that introduces genetic diversity via the process of recombination and independent assortment. Following the fusion of male and female gametes, the combination of paternal and maternal genetic material in the zygote allows for genetic recombination, which is a fundamental source of genetic variation and, thus, evolution. Therefore, gametes not only are critical to the inception of new individuals but also contribute to species survival and evolution.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000300") -AnnotationAssertion(Annotation( "DOI:10.1016/j.jri.2023.103811") Annotation( "DOI:10.1530/JOE-17-0402") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK53245") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.jri.2023.103811") Annotation( "DOI:10.1530/JOE-17-0402") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK53245") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Trophoblast cells, originating from the blastocyst's outer layer, are highly specialized and pivotal for embryo development. Their crucial roles include facilitating implantation and contributing to the formation of the placenta, an essential organ for fetal nutrition, gas exchange, and waste removal during pregnancy. These cells play a crucial role in implantation by adhering to the uterus lining. Post-implantation, some trophoblast cells differentiate, forming syncytiotrophoblasts. These multi-nucleated cells absorb nutrients, release waste, and secrete hormones, including human chorionic gonadotropin (hCG), sustaining pregnancy. Trophoblast cells also play a crucial role in immune tolerance, preventing the maternal immune system from treating the fetus as foreign. This balance is vital for a healthy pregnancy. Dysfunctional trophoblast cells may lead to complications like miscarriages or preeclampsia, emphasizing their importance in both embryology and reproductive medicine.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000351") -AnnotationAssertion(Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK482438/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532857/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK557452/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK482438/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532857/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK557452/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Smooth muscle cells, as a broader category, are non-striated, involuntary muscle cells that usually exhibit contractions and dilation to facilitate numerous physiological functions. Sphincter associated smooth muscle cells are a specific type of smooth muscle cell that are primarily associated with the functioning of various anatomical sphincters in the human body. The traits that distinguishes sphincter associated smooth muscle cells from other smooth muscle cells are their location and specialized function in sphincter mechanisms. Sphincter associated smooth muscle cells play a pivotal role in regulating the opening and closure of the bodily sphincters. These sphincters can be found at multiple locations within the body including the gastrointestinal tract (namely the esophagus, stomach, small intestine, and rectum), the urinary tract, and the circulatory system. Upon receiving signals from the autonomous nervous system, these cells contract or relax to modulate the aperture of sphincters, thus controlling the movement of substances through them. Due to their precise and vital functional role, any dysfunction or dysregulation in these cells can lead to serious medical conditions, such as gastroesophageal reflux or urinary incontinence.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000358") -AnnotationAssertion(Annotation( "DOI:10.1093/cvr/cvs135") Annotation( "DOI:10.1093/cvr/cvy023") Annotation( "DOI:10.1161/ATVBAHA.118.311229") Annotation( "DOI:10.3389/fimmu.2020.599415") Annotation( "DOI:10.3390/ijms20225694") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1093/cvr/cvs135") Annotation( "DOI:10.1093/cvr/cvy023") Annotation( "DOI:10.1161/ATVBAHA.118.311229") Annotation( "DOI:10.3389/fimmu.2020.599415") Annotation( "DOI:10.3390/ijms20225694") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Vascular associated smooth muscle cells (VSMCs) are specialized cells that constitute a major component of the blood vessel wall in the circulatory system. They derive their name from the predominantly smooth appearance under microscopic examination, as opposed to the striated appearance of cardiac and skeletal muscles. A principal function of VSMCs is to regulate vascular tone, which directly dictates blood pressure and blood flow distribution across various body parts. This is achieved through coordinated contraction and relaxation of the muscles in response to a variety of extracellular signals. When a blood vessel is exposed to stimuli like pressure or injury, VSMCs contract, resulting in vasoconstriction and ultimately, an increase in blood pressure. Conversely, in response to vasodilators, these cells relax, leading to vasodilation and a consequent decrease in blood pressure. VSMCs are also involved in vascular remodeling, a process where blood vessels adapt their structure to long-term changes in hemodynamic conditions or disease states. In diseases like atherosclerosis and hypertension, an aberrant proliferative and synthetic phenotype of VSMCs contributes to the formation of plaques, leading to vessel hardening or even occlusion. Recent research has further implicated these cells in immune responses, recognizing their function in leukocyte recruitment and inflammation, factors that are largely instrumental in vascular pathologies.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000359") -AnnotationAssertion(Annotation( "DOI:10.1016/j.semcdb.2016.04.009") Annotation( "DOI:10.1242/dev.202046") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK26843/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553142/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.semcdb.2016.04.009") Annotation( "DOI:10.1242/dev.202046") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK26843/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553142/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The male gamete, also known as a sperm cell, is a highly specialized cell type critical to sexual reproduction. Sperm are produced during a process called spermatogenesis within the male reproductive organ, specifically the testes. They are haploid cells, carrying the genetic material from the male parent, and fuse with the ovum, the female gamete, to form a diploid zygote during fertilization, thereby initiating gestation and the development of a new organism. The male gamete is distinguished by its morphology, notably its elongated shape which is geared towards locomotion. One of the primary characteristics of a male gamete is its flagellum, or tail, which propels the cell towards the female gamete. It additionally contains an elongated head region housing the nucleus, where the genetic material is stored. The anterior section of the head, the acrosome, releases enzymes essential for penetrating the protective layers of the ovum during the fertilization process. The function of male gametes does not cease upon fertilization. In addition to delivering the paternal set of chromosomes, fusion of the sperm cell with the ovum causes an increase in cytosolic Ca2+ which then activates the female egg cell to undergo the so-called cortical reaction during which the egg’s zona pellucida is changed so fusion with additional sperm is prevented. The Ca2+ signal also initiates the development of the zygote and eventual formation of an embryo. Male gametes also contribute towards forming the placenta, which supplies nutrients and removes waste products during gestation.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000408") -AnnotationAssertion(Annotation( "DOI:10.1016/j.ejogrb.2004.01.010") Annotation( "DOI:10.1093/humrep/del408") Annotation( "DOI:10.1093/humupd/6.3.279") Annotation( "DOI:10.1210/jcem-28-3-355") Annotation( "DOI:10.3389/fendo.2019.00832/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.ejogrb.2004.01.010") Annotation( "DOI:10.1093/humrep/del408") Annotation( "DOI:10.1093/humupd/6.3.279") Annotation( "DOI:10.1210/jcem-28-3-355") Annotation( "DOI:10.3389/fendo.2019.00832/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Granulosa cells are a type of somatic cell most commonly known for their crucial role within the ovarian follicles of female mammalian species. Named for their grainy appearance, they are situated in the follicular epithelium, lining the inner part of the follicle and directly surrounding the oocyte. These cells stand as an integral part of the ovarian structure and function. One of the primary roles of granulosa cells is to aid in the production and secretion of sex hormones, particularly estrogen. They achieve this by working in tandem with theca cells, which stay attached to the external layer of the follicle. Theca cells produce androstenedione (a type of androgen) which granulosa cells then convert into estradiol, a form of estrogen, with the help of the enzyme aromatase. Moreover, granulosa cells participate in luteinization, transforming into luteal granulosa cells as a response to the luteinizing hormone during ovulation. This allows the formation of the corpus luteum, responsible for the secretion of progesterone necessary to maintain pregnancy. Granulosa cells also play a significant role in follicular development and oocyte maturation, involving close communication with the contained oocyte. They support the oocyte through the provision of nutrients and growth factors, control its meiotic cycle and ensure it is appropriately oriented and instructed for impending ovulation. Furthermore, granulosa cells contribute to the formation of the zona pellucida and the follicular fluid, providing an optimal environment for the oocyte's growth and maturation. These cells, thus, perform multiple vital roles, underscoring their importance in fertility and reproductive health.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000501") -AnnotationAssertion(Annotation( "DOI:10.1016/j.pharmthera.2015.05.007") Annotation( "DOI:10.1038/s41574-018-0020-6") Annotation( "DOI:10.1093/annonc/mdh216") Annotation( "https://www.sciencedirect.com/topics/neuroscience/somatostatin-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.pharmthera.2015.05.007") Annotation( "DOI:10.1038/s41574-018-0020-6") Annotation( "DOI:10.1093/annonc/mdh216") Annotation( "https://www.sciencedirect.com/topics/neuroscience/somatostatin-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Type D enteroendocrine cells, also known as D or delta cells, are specialized hormone-releasing cells found in the pancreas and also scattered throughout the lining of the gastrointestinal tract in mammals, notably within the stomach and the upper part of the small intestine known as the duodenum. The primary role of D cells is to produce and secrete somatostatin, a potent paracrine inhibitor. In the gastrointestinal tract, somatostatin slows down digestion. It reduces gastric acid secretion and slows down the rate of gastric emptying, thereby prolonging and controling the digestive p. Functionally, these effects are aimed at sustaining nutrient absorption to optimize energy extraction from consumed food. In the pancreas, D cells maintain a vital role in endocrine regulation. D cells in the pancreatic islands secrete somatostatin to inhibit the release of both insulin and glucagon from type A cells and B cells, glucoregulatory hormones that control blood sugar levels. Hence, D cells contribute considerably to the homeostasis of the body's metabolic processes. It is also noteworthy that dysfunctional D cells or irregular somatostatin signaling has been associated with certain pathologies such as neuroendocrine tumors and gastric ulcers.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000502") -AnnotationAssertion(Annotation( "DOI:10.1007/s00018-002-8412-z") Annotation( "DOI:10.1038/nature09637") Annotation( "DOI:10.1038/nrmicro2546") Annotation( "DOI:10.1146/annurev-physiol-030212-183744") Annotation( "DOI:10.3389/fimmu.2020.00587") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00018-002-8412-z") Annotation( "DOI:10.1038/nature09637") Annotation( "DOI:10.1038/nrmicro2546") Annotation( "DOI:10.1146/annurev-physiol-030212-183744") Annotation( "DOI:10.3389/fimmu.2020.00587") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Paneth cells are specialized epithelial cells that are primarily located at the bottom of the crypts of Lieberkühn in the small intestine, where they play a pivotal role in maintaining gut homeostasis. They have also been found in smaller numbers in the colonic crypts and other parts of the gastrointestinal tract. Paneth cells are characterized by large acidophilic granules, which take up most of the cytoplasmic volume, causing the nucleus to be pushed toward the base of the cell. Paneth cells function as a part of the innate immune system. The large granules inside the cells are filled with antimicrobial peptides, such as defensins and lysozymes. Upon bacterial intrusion or cellular signaling indicating a potential infection, Paneth cells release the granules containing the antimicrobial substances into the crypt lumen, effectively serving as the first line of defense against bacterial invasion within the gastrointestinal tract. In essence, Paneth cells serve as guardians, protecting the intestinal stem cells from harmful pathogens that may disturb the gut ecosystem. In addition to their primary role in immunity, Paneth cells are also crucial for supporting the stem cell niche in the intestinal crypts. They are located adjacent to Lgr5+ stem cells and secrete various growth factors such as EGF, TGF-alpha, Wnt3, and Notch ligand Dll4. These factors regulate the self-renewal and differentiation of these stem cells, which continuously replenish the intestinal epithelium. Consequently, any abnormality or dysfunction in Paneth cells could lead to a disturbance in gut homeostasis, possibly resulting in various illnesses such as inflammatory bowel disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000510") -AnnotationAssertion(Annotation( "DOI:10.1074/jbc.RA118.001739") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK544225/") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/smooth-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1074/jbc.RA118.001739") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK544225/") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/smooth-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. A precursor to the smooth muscle cell, the smooth muscle myoblast, is integral to various body systems, including the cardiovascular, gastrointestinal, and respiratory systems. Smooth muscle myoblasts arise from the differentiation of mesenchymal stem cells, a process tightly regulated by specific genetic factors and environmental signals. Their role extends to myogenesis, particularly in the formation of muscular tissue, especially during embryonic development. Smooth muscle myoblasts possess distinctive cellular characteristics. In contrast to skeletal muscle myoblasts, they do not fuse to create multinucleated fibers. Instead, they differentiate into individual, uninucleated smooth muscle cells. This differentiation unfolds in two distinct phases: proliferation, marked by rapid division to increase cell numbers, and differentiation, involving structural and functional transformation into mature smooth muscle cells. This intricate process is intricately mediated by various signaling molecules and transcription factors. Functionally, smooth muscle myoblasts and the mature muscle cells they generate play pivotal roles in various physiological processes. Smooth muscle cells orchestrate involuntary muscle responses and are integral to body systems that regulate blood flow, propel food along the digestive tract, and control airflow in the respiratory tract. These diverse functions underscore the significance of smooth muscle myoblasts in the body's growth, maintenance, and overall function.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000514") -AnnotationAssertion(Annotation( "DOI:10.1016/j.immuni.2017.10.021") Annotation( "DOI:10.1146/annurev-immunol-081022113627") Annotation( "DOI:10.1186/2050-7771-2-1") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.immuni.2017.10.021") Annotation( "DOI:10.1146/annurev-immunol-081022113627") Annotation( "DOI:10.1186/2050-7771-2-1") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Granulocyte monocyte progenitor cells (GMP cells), originating from hematopoietic stem cells in the bone marrow, are a critical intermediate in myeloid cell development. Positioned at a key differentiation juncture, these cells exhibit bidirectional potential, giving rise to both granulocyte and monocyte lineages. Their primary role is to generate mature granulocytes and monocytes, essential components of the body's innate immunity and inflammatory responses. Granulocytes, such as neutrophils, eosinophils, and basophils, contribute to the first line of defense against infections by migrating to the infection site and phagocytosing pathogens. Monocytes can differentiate into macrophages or dendritic cells, playing diverse roles in immune responses.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000557") -AnnotationAssertion(Annotation( "DOI:10.1016/s1357-2725(03)00042-6") Annotation( "DOI:10.1152/physrev.00061.2017") Annotation( "DOI:10.1186/s13287-022-02706-5") Annotation( "DOI:10.1369/0022155411426780") Annotation( "DOI:10.22074/cellj.2016.4714") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/s1357-2725(03)00042-6") Annotation( "DOI:10.1152/physrev.00061.2017") Annotation( "DOI:10.1186/s13287-022-02706-5") Annotation( "DOI:10.1369/0022155411426780") Annotation( "DOI:10.22074/cellj.2016.4714") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Skeletal muscle satellite cells (SMSCs) are a type of adult stem cell that nestles on the outer surface of the myofiber, positioned between the sarcolemma (plasma membrane of the muscle cell) and peripheral basement membrane. The primary function of SMSCs lies in their capacity to facilitate the growth and repair of damaged skeletal muscle. Upon muscle injury or trauma, these normally quiescent cells activate, proliferate, and differentiate into myoblasts. These myoblasts subsequently mature and fuse to form myotubes, which ultimately help in the repair and rebuilding of muscle fibers. This response is an integral part of the skeletal muscle's ability to recover from injury and adapt to enhanced or changing physical demands, thereby maintaining and improving muscle function. Furthermore, SMSCs also contribute to hypertrophic muscle growth due to resistance exercise and endurance training, with the notable feature of their multi-nucleated nature resulting from the fusion of differentiated myoblasts. The additional nuclei support enhanced protein synthesis, thus allowing the muscle fibers to grow and strengthen in response to exercise. In aging and in diseases, the number or functionality of these cells can decrease, leading to impaired muscle regeneration and progressive muscle weakness.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000594") -AnnotationAssertion(Annotation( "DOI:10.1007/BF00710764") Annotation( "DOI:10.1038/nrgastro.2013.36") Annotation( "DOI:10.1097/MOG.0b013e32832ebfac") Annotation( "DOI:10.1111/prd.12116") Annotation( "DOI:10.1152/physrev.00011.2011") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/BF00710764") Annotation( "DOI:10.1038/nrgastro.2013.36") Annotation( "DOI:10.1097/MOG.0b013e32832ebfac") Annotation( "DOI:10.1111/prd.12116") Annotation( "DOI:10.1152/physrev.00011.2011") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Acinar cells are specialized exocrine gland cells that secrete specific enzymes and fluids to aid in digestion. Found primarily in the pancreas and salivary glands, the term acinar is derived from the Latin word 'acinus' which means 'grape'; this is because acinar cells are arranged in a grape-like cluster around small ducts, formulating an acinus structure. In the salivary glands, acinar cells secrete digestive enzymes as well as other substances such as mucus and water. These secretions service the initial stages of the digestive process, preparing the consumed food for onward digestion in the stomach and intestines by lubricating and partially breaking it down. In the pancreas, acinar cells are responsible for synthesizing and secreting a significant amount of digestive enzymes, such as trypsin, chymotrypsin, and amylase. These enzymes are stored in zymogen granules inside the acinar cells until they are dispatched into the small intestine. Once within the small intestine, they break down proteins, carbohydrates, and fats into substances that can be absorbed. This is vital to the effective and efficient digestion and absorption of nutrients from the food we consume. Acinar cells have high protein synthesis rates and are susceptible to accumulation of misfolded proteins; the subsequential induction of ER stress is thought to be involved in the development of pancreatitis, a serious inflammatory disease of the exocrine pancreas.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000622") -AnnotationAssertion(Annotation( "DOI:10.1038/s41467-022-33748-1") Annotation( "DOI:10.1038/s41572-020-0196-7") Annotation( "DOI:10.1146/annurev-physiol-020911-153238") Annotation( "DOI:10.1159/000481633") Annotation( "DOI:10.3389/fcell.2021.771931") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41467-022-33748-1") Annotation( "DOI:10.1038/s41572-020-0196-7") Annotation( "DOI:10.1146/annurev-physiol-020911-153238") Annotation( "DOI:10.1159/000481633") Annotation( "DOI:10.3389/fcell.2021.771931") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Podocytes are highly specialized, terminally differentiated glomerular visceral epithelial cells that wrap around the capillaries in the kidneys. They play an essential role in kidney function, particularly in the filtration selectivity of the glomerulus. Each podocyte is characterized by a unique architecture with a large cell body, ‘major processes’ extending outwardly from the cell body and ‘foot processes’, also known as pedicels, which surround. the glomerular capillary loops Podocytes represent the last barrier of the glomerular filtration membrane in the kidney. This barrier prevents the leakage of plasma proteins from the blood into the urine, hence maintaining protein homeostasis in the body. The foot processes of the podocytes interdigitate with those from neighboring cells. The cell-cell junctions between the foot processes, the slit-diaphragms, are thought to create a molecular sensor of renal filtration that prevents the passage of macromolecules while allowing water and small solutes to pass. Podocytes also contribute to the glomerular basement membrane by secreting collagen and maintain glomerular endothelial cell fenestration by secreting VEGFA. They have been shown to play a role in inducing cytoskeletal regulation, cell adhesion, and inflammatory response, consistent with their essential function in the kidney. The importance of podocytes is further emphasized by the effects of their damage or loss. Abnormalities in podocytes often result in severe kidney diseases (podocytopathies), including focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD). Injury to the podocytes can result in \"effacement\" or flattening of foot processes, leading to increased permeability of the filtration barrier and proteinuria (an excess of serum protein in urine) which is a common symptom of kidney diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000653") -AnnotationAssertion(Annotation( "DOI:10.1002/ar.1092200109") Annotation( "DOI:10.1016/0306-4522(86)90162-4") Annotation( "DOI:10.1016/j.devcel.2012.11.003") Annotation( "DOI:10.1038/s41467-022-31571-2") Annotation( "DOI:10.1152/ajprenal.90601.2008") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/ar.1092200109") Annotation( "DOI:10.1016/0306-4522(86)90162-4") Annotation( "DOI:10.1016/j.devcel.2012.11.003") Annotation( "DOI:10.1038/s41467-022-31571-2") Annotation( "DOI:10.1152/ajprenal.90601.2008") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Fenestrated cells are specialized epithelial cells that are characterized by a distinctive structural feature, fenestrations or tiny pores, which allow an exchange of substances such as fluids, nutrients, and waste between blood vessels and the surrounding tissue environments. These cells are present in various types of epithelial tissues in the human body, including the intestinal tract, endocrine glands, and certain parts of the renal system. There are multiple types of endothelial cell fenestrations. The most common type is found in systemic capillaries of the endocrine tissue (e.g., pancreatic islets), gastrointestinal mucosa, and renal peritubular capillaries. Here, the fenestrated cells have fenestrations in their peripheral cytoplasm with a unique thin and permeable diaphragm that provides a high degree of selectivity, blocking the passage of larger molecules while allowing the free movement of smaller ones. In contrast, other types of fenestrations, such as the discontinuous epithelium of the liver sinusoidal endothelial cells, do not have diaphragms. @@ -474,570 +453,570 @@ Characteristically, fenestrated cells play a vital role in the filtering process In the endocrine system, fenestrated cells in glandular capillaries allow the prompt release of hormones into the bloodstream. These cells have tightly clustered fenestrations which increase the surface area available for passive diffusion, improving the efficiency of hormone secretion. In villi of the small intestine, these cells increase absorption efficiency, permitting the exchange of water, electrolytes, and nutrients. Overall, fenestrated cells contribute immensely to important physiological processes of filtration, absorption, and secretion, primarily connecting our body's circulation system with the surrounding tissues and organs.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000666") -AnnotationAssertion(Annotation( "DOI:10.1038/nature08529") Annotation( "DOI:10.1093/jb/mvv121") Annotation( "DOI:10.1136/gut.47.5.735") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK534232/") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/microfold-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nature08529") Annotation( "DOI:10.1093/jb/mvv121") Annotation( "DOI:10.1136/gut.47.5.735") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK534232/") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/microfold-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. M cells, or microfold cells, of the gut are specialized epithelial cells found in the lining of the gut, specifically in the follicle-associated epithelium of mucosa-associated lymphoid tissue, where they function as sentries against toxins and pathogens. M cells are characterized by apical microfolds (hence their alternate name) which express unique adhesion molecules that enable them to sample the luminal macromolecules. Other morphological features that distinguish M cells from other intestinal mucosal cells include sparse microvilli and a reduced thickness of the glycocalyx, which permits adhesion while not hindering the transport of molecules. They also have unique intraepithelial invaginations on the basolateral side which are filled with macrophages and other immune cells that can process the engulfed macromolecules quickly. These morphological characteristics enable M cells to serve a dual role in immune responses. They initiate the immune response by transporting antigens (such as toxic or pathogenic substances) across the epithelial layer to lymphocytes and antigen-presenting cells in the underlying lymphoid tissue. This specialized transport process is called 'transcytosis.' They also have specialized molecules like glycoprotein-2 for bacterial uptake. Simultaneously, M cells help maintain immune tolerance to food antigens and commensal bacteria, preventing unnecessary reactions to non-pathogenic substances and hypersensitivity conditions. While these functions are crucial for well-being, M cell dysfunction can lead to serious conditions like Crohn's disease and other inflammatory bowel diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000682") -AnnotationAssertion(Annotation( "DOI:10.1016/j.febslet.2014.07.005") Annotation( "DOI:10.1016/j.mce.2015.06.028") Annotation( "DOI:10.1038/s41580-020-00317-7") Annotation( "DOI:10.1210/jc.2003-030630") Annotation( "DOI:10.1369/00221554155835") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.febslet.2014.07.005") Annotation( "DOI:10.1016/j.mce.2015.06.028") Annotation( "DOI:10.1038/s41580-020-00317-7") Annotation( "DOI:10.1210/jc.2003-030630") Annotation( "DOI:10.1369/00221554155835") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. PP cells, also known as pancreatic polypeptide cells (and previously called F cells or gamma cells), are enteroendocrine cells predominantly found in the islets of Langerhans in the head of the pancreas. They are one of the four main endocrine cell types present in the pancreatic islets, along with type A, B and D cells. PP cells are notable for their production of pancreatic polypeptide, an anorexigenic hormone that modulates food intake and energy homeostasis. By secreting pancreatic polypeptide, PP cells play a significant role in the management of both digestive and appetite regulation. Upon ingestion of food, there is a significant increase in the secretion of pancreatic polypeptide, which then reduces biliary secretion and helps slow down the movement of food through the digestive tract. This allows more time for digestion to take place and nutrients to be absorbed, promoting the efficient use of dietary intake. The pancreatic polypeptide further reduces appetite by interacting with the hypothalamus, the area of the brain responsible for control of hunger. Given their important role in digestion, malfunction or damage to PP cells can lead to a disturbance in the digestive process and contribute to some disease conditions. For example, an overproduction of pancreatic polypeptide can result in conditions such as pancreatic tumors and diabetes. Conversely, an under secretion might contribute to obesity due to impaired dietary control. Furthermore, PP cells may also play a role in the body's energy balance, suggesting their implication in conditions related to energy metabolism.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000696") -AnnotationAssertion(Annotation( "DOI:10.1002/nau.22195") Annotation( "DOI:10.1038/ki.2009.73") Annotation( "DOI:10.1038/ncpuro0672") Annotation( "DOI:10.1038/s41579-020-0324-0") Annotation( "DOI:10.1152/physrev.00041.2019") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/nau.22195") Annotation( "DOI:10.1038/ki.2009.73") Annotation( "DOI:10.1038/ncpuro0672") Annotation( "DOI:10.1038/s41579-020-0324-0") Annotation( "DOI:10.1152/physrev.00041.2019") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Urothelial cells are a unique type of epithelial cell found lining the urinary tract system. They form the urothelium, a specialized, multi-layered epithelium that lines major portions of the urinary tract, including the renal pelvis, ureters, bladder, and the proximal part of the urethra. The distinct characteristic of urothelial cells is their ability to stretch and contract depending on the volume of liquid they contain - a feature that facilitates the essential role they play in maintaining the functionality and integrity of the urinary system. The primary function of urothelial cells is to provide an impermeable barrier to urine, preventing the toxic components present in the urine from seeping back into the body's bloodstream. The urothelium consists of a superficial umbrella cell layer, 1–2 layers of intermediate cells, and a basal cell layer. The umbrella layer is composed of large, mostly binucleated cells that are covered in an asymmetrical unit membrane, the uroplakin plaques. These plaques function to seal the apical membrane of the bladder from the toxic and highly variable contents of urine. The lipid bilayer structure of uroplakin plaques has an exceptionally high concentration of uroplakins that contribute to the barrier function of the urothelium. Beyond their mechanical function, urothelial cells also play a role in sensing and signaling changes in the urinary system. They express a number of sensor molecules or respond to thermal, mechanical and chemical stimuli and can release chemical mediators. This allows them to act as sensory transducers because they detect changes in the filling state of the bladder and transmit this information to the nervous system. Moreover, these versatile cells contribute to the defense mechanism against urinary tract pathogens. They respond to bacterial infections by releasing chemical messengers, cytokines and chemokines, to attract immune cells, and also can engulf pathogens via endocytosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000731") -AnnotationAssertion(Annotation( "DOI:10.1016/j.ccep.2010.10.012") Annotation( "DOI:10.1038/nrcardio.2016.203") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK572070") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/cardiac-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.ccep.2010.10.012") Annotation( "DOI:10.1038/nrcardio.2016.203") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK572070") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/cardiac-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Cardiac muscle cells, also known as cardiomyocytes or cardiac myocytes, are specialized cells that form the heart tissue. These cells are elongated, branched, and contain a single centrally located nucleus. Their anatomy is composed primarily of densely packed myofibrils, which are protein structures that consist of sarcomeres - the fundamental units of muscle contraction. Cardiac muscle cells are united at their ends through specialized junctions known as intercalated discs, which allow the heart to contract in a unified, powerful and rhythmic way. Functionally, cardiac muscle cells are responsible for the heart's consistent pumping action that circulates blood throughout the body. Unlike most cells in the body, cardiac muscle cells spontaneously depolarize and generate action potentials without external stimulation. This unique trait stems from the presence of ion channels in the cells' membrane that allow a cyclic flow of ions across the membrane, which create the electrical impulses necessary for heart contraction. The spread of these electrical signals from one cardiac muscle cell to another - facilitated by the interconnected network made by the intercalated disks - results in a synchronized contraction of the heart muscle. Unlike skeletal muscle cells which can tire and need rest, cardiac muscle cells have to work ceaselessly throughout the entire lifespan, without the opportunity for rest, to ensure continuous circulation of blood. This is made possible through the high volume of mitochondria and a constant supply of oxygen from coronary circulation. In conclusion, cardiac muscle cells, through their unique structure and vital functionality, play a pivotal role in sustaining life by providing the means for blood to reach every cell in the body.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000746") -AnnotationAssertion(Annotation( "DOI:10.1038/nri.2017.28") Annotation( "DOI:10.1111/sji.12883") Annotation( "DOI:10.1182/blood-2009-07-235028") Annotation( "DOI:10.3389/fimmu.2015.00423/full") Annotation( "DOI:10.3389/fimmu.2019.02035") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nri.2017.28") Annotation( "DOI:10.1111/sji.12883") Annotation( "DOI:10.1182/blood-2009-07-235028") Annotation( "DOI:10.3389/fimmu.2015.00423/full") Annotation( "DOI:10.3389/fimmu.2019.02035") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Classical monocytes are a subtype of monocytes that are characterized by high CD14 but no CD16 expression. Emerging from the bone marrow and entering the bloodstream, these cells play central roles in immune responses and regulation of inflammation. CD14-positive CD16-negative monocytes form the majority of circulating monocytes in the body, typically contributing to around 80-90% of the total monocyte pool. The primary function of the classical monocytes is to serve in the frontline of host defense against infections. They are primed to migrate to sites of infection, and they express pattern recognition receptors that help them identify and phagocytose pathogens, leading to their destruction. Classical monocytes also contribute to inflammation by producing several pro-inflammatory cytokines including interleukins and tumor necrosis factors. In response to specific signals from tissues under pathological conditions, such as infection or injury, classical monocytes can leave the bloodstream and migrate towards the affected sites. Following their arrival, these cells differentiate into diverse cell types including macrophages and dendritic cells to combat specific pathogens or injury. Dysregulated monocyte activity can lead to the development of many human diseases including inflammation, infection, tissue injury, and various autoimmune diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000860") -AnnotationAssertion(Annotation( "DOI:10.1038/s41392-023-01452-1") Annotation( "DOI:10.1186/s12935-021-02089-2") Annotation( "DOI:10.3389/fimmu.2015.00263") Annotation( "DOI:10.3389/fimmu.2020.583084") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41392-023-01452-1") Annotation( "DOI:10.1186/s12935-021-02089-2") Annotation( "DOI:10.3389/fimmu.2015.00263") Annotation( "DOI:10.3389/fimmu.2020.583084") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Inflammatory macrophages, also sometimes referred to as M1 or classically activated macrophages, play an important role in the inflammatory response. The M1/M2 classification is based upon macrophage polarization rather than macrophage location and refers to macrophage activation towards either a more inflammatory or more resolving phenotype, respectively, although the functional diversity of macrophages is more nuanced than such a dichotomy suggests. Inflammatory macrophages are derived from monocytes recruited to a site of infection or injury. M1 macrophages are classically activated, typically by IFN-γ or lipopolysaccharide (LPS). Upon sensing signs of inflammation, they quickly respond by increasing their pro-inflammatory activity. They achieve this by producing a range of signaling molecules, such as nitric oxide, reactive oxygen species, and numerous cytokines such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-1, -6 and -12. The release of these potent molecules helps to recruit other immune cells to the site, killing off pathogenic organisms and facilitation inflammation. At the same time, M1 macrophages can also present antigens to T cells, thereby helping to induce an adaptive immune response. Over time, these macrophages may transition in phenotype and function to help resolve the inflammation and promote tissue repair. Despite the beneficial role of inflammatory macrophages in dealing with pathogens, chronic activation of these cells can lead to harmful effects. Over time, continuous production of pro-inflammatory molecules can cause damage to tissues and organs. This is seen in certain chronic inflammatory diseases, such as atherosclerosis, diabetes, obesity, asthma, and various autoimmune disorders. In such situations, the normally protective function of inflammatory macrophages is not properly controlled, which can contribute to disease pathology.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000863") -AnnotationAssertion(Annotation( "DOI:10.1007/s00281-013-0382-8") Annotation( "DOI:10.1016/j.immuni.2022.10.005") Annotation( "DOI:10.1016/j.tins.2021.07.002") Annotation( "DOI:10.1038/s41583-019-0201-x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00281-013-0382-8") Annotation( "DOI:10.1016/j.immuni.2022.10.005") Annotation( "DOI:10.1016/j.tins.2021.07.002") Annotation( "DOI:10.1038/s41583-019-0201-x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Central Nervous System (CNS) macrophages represent an integral part of the brain's innate immune system. These immune cells play crucial roles in maintenance and regulation, homeostasis, and disease response, thereby helping in the overall cognitive functioning of an organism. There are different macrophage populations in the CNS, often classified into microglial cells, which reside in the parenchyma, and non-parenchymal macrophages at the interface between the brain and the periphery, including the perivascular spaces, the choroid plexus, and the meninges. CNS macrophages are derived from erythromyeloid progenitors in the yolk sac. CNS macrophages have distinct morphologies: Meningeal and perivascular macrophages have a more elongated shape than microglia, which are characterized by a small cell body with fine processes; in contrast, choroid plexus macrophages typically have a stellate shape. Microglia are the most abundant abundant mononuclear phagocytes and have been shown to play a number of physiological roles, including proinflammatory and anti-inflammatory functions, synaptic pruning and remodeling, and apoptotic cell removal through phagocytosis. Non-parenchymal CAMs primarily support the barrier function against external antigens. Meningeal macrophages have been shown to respond to peripheral microbial challenges and to protect the brain against fatal viral infection. CNS macrophages are also involved in the initiation and progression of many neurological diseases, such as Alzheimer's disease and Parkinson's disease as well as multiple sclerosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000878") -AnnotationAssertion(Annotation( "DOI:10.1038/s41392-023-01452-1") Annotation( "DOI:10.1186/s12935-021-02089-2") Annotation( "DOI:10.3389/fimmu.2015.00263") Annotation( "DOI:10.3389/fimmu.2020.583084") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41392-023-01452-1") Annotation( "DOI:10.1186/s12935-021-02089-2") Annotation( "DOI:10.3389/fimmu.2015.00263") Annotation( "DOI:10.3389/fimmu.2020.583084") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Alternatively activated macrophages, also referred to as M2 macrophages, are immune cells originating from monocytes. The M1/M2 classification is based upon macrophage polarization rather than macrophage location and refers to macrophage activation towards either a more inflammatory or more resolving phenotype, respectively, although the functional diversity of macrophages is more nuanced than such a dichotomy suggests. M2 macrophages cells are differentiated from their precursors generally in response to Th2 cytokines, such as interleukin-4 (IL-4), interleukin-10 (IL-10), and interleukin-13 (IL-13). Tissue-resident macrophages are also sometimes said to have an “M2-like” phenotype. M2 macrophages play key roles in immunoregulation and disease resolution. Functionally, alternatively activated macrophages are essential in wound healing and tissue repair and remodeling, largely due to their potent anti-inflammatory actions and their ability to promote angiogenesis. They achieve these functions by the production of specific growth factors and signaling proteins including Arg1, Ym1/2, Fizz1, and TGF-β. Additionally, they provide defense against specific categories of pathogens, particularly parasites, through specific communication with Th2 cells. However, dysfunction of M2 macrophages can be harmful. Alternatively activated macrophages have been associated with several pathological conditions such as asthma, fibrosis, and tumor progression. This is due to their capacity to inhibit inflammatory responses, promote unneeded wound healing processes, and support tumour growth and spreading. Furthermore, complex roles have been observed in metabolic disorders, cardiovascular diseases, and neurodegenerative diseases, showing the diverse functional spectrum of these cells.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0000890") -AnnotationAssertion(Annotation( "DOI:10.1007/s00795-015-0099-y") Annotation( "DOI:10.1007/s11914-012-0105-4") Annotation( "DOI:10.1016/j.jot.2021.04.005") Annotation( "DOI:10.1038/s41413-020-0099-y") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00795-015-0099-y") Annotation( "DOI:10.1007/s11914-012-0105-4") Annotation( "DOI:10.1016/j.jot.2021.04.005") Annotation( "DOI:10.1038/s41413-020-0099-y") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Bone cells, also known as osteocytes, form the building blocks of the skeletal system. They represent the most common type of bone cell, making up approximately 95% of the total bone cell population in mature adult bone tissue. Osteocytes are a stellate shape with numerous long, slender dendritic processes. The formation of bone is a complex process involving the action of osteoclasts - cells that break down and reabsorb old bone, followed by osteoblasts that rebuild the bone. Osteocytes serve as coordinators for these two types of bone cells to ensure a balance in the bone remodeling process, contributing to maintaining both bone strength and mineral homeostasis. The primary functions of osteocytes involve the creation, maintenance, and repair of bone tissue. They ensure that bone remodeling is performed by forming a network within the bone and communicating with other cells to help them respond to mechanical strain and damage. A remarkable aspect of osteocytes is their ability to perceive mechanical forces, translating them into biochemical signals that regulate bone remodeling and adaptation. Defects in osteocyte function, aside from other bone cells, have been associated with several bone disorders such as osteoporosis, renal osteodystrophy, and skeletal manifestations of diabetes.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0001035") -AnnotationAssertion(Annotation( "DOI:10.1016/0165-6147(89)90192-2") Annotation( "DOI:10.1038/nrgastro.2013.36") Annotation( "DOI:10.1097/01.mog.0000239863.96833.c0") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK54134/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/0165-6147(89)90192-2") Annotation( "DOI:10.1038/nrgastro.2013.36") Annotation( "DOI:10.1097/01.mog.0000239863.96833.c0") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK54134/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Pancreatic acinar cells are the functional units of the exocrine pancreas. These cells are structurally arranged in small clusters - or acini - with a small central lumen, and their primary function is to produce and secrete enzymes that facilitate digestion of proteins, carbohydrates, and fats within the small intestine. Some of the most important digestive enzymes synthesized by the pancreatic acinar cells include amylase, which digests carbohydrates, lipase, which breaks down fats, and several proteases such as trypsin and chymotrypsin which are vital for the digestion of proteins. Importantly, these enzymes are produced as proenzymes or zymogens to avoid autolysis, or the breakdown of the pancreas itself. The release of these enzymes by the pancreatic acinar cells is strictly regulated by hormones and neurochemicals. Cholecystokinin (CCK) released from the enteroendocrine cells in the duodenum cause the release of these enzymes. Moreover, acinar cells also respond to another hormone, secretin, by releasing bicarbonate-rich fluid which helps to neutralize the gastric acid in the duodenum. Dysregulation in the function of pancreatic acinar cells can lead to severe health issues, including pancreatitis, which is characterized by inflammatory damage to the organ due to the premature activation of digestive enzymes.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002064") -AnnotationAssertion(Annotation( "DOI:10.1016/j.diabres.2018.06.013") Annotation( "DOI:10.1210/en.2016-1748") Annotation( "DOI:10.2337/db15-1541") Annotation( "DOI:10.2337/dbi19-0002") Annotation( "DOI:10.3389/fphys.2012.00349/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.diabres.2018.06.013") Annotation( "DOI:10.1210/en.2016-1748") Annotation( "DOI:10.2337/db15-1541") Annotation( "DOI:10.2337/dbi19-0002") Annotation( "DOI:10.3389/fphys.2012.00349/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Type A enteroendocrine cells, also known as alpha cells or A cells, are a species of endocrine cells primarily located in the pancreatic islets of Langerhans; they have also been identified within the lining of the stomach. Functionally, these cells are pivotal in glucose metabolism and homeostasis, accounting for about 20% of the total population of cells in the pancreatic islets. The primary role of type A enteroendocrine cells involves the synthesis, storage, and secretion of the peptide hormone glucagon, which is critical in energy regulation throughout the body. In response to a decrease in blood glucose levels, the pancreatic A cells are stimulated to secrete glucagon into the bloodstream. Glucagon acts on its target cells, mainly in the liver, to stimulate glycogenolysis and gluconeogenesis processes, thereby increasing blood glucose levels back to normal. In this way, pancreatic A cells play an integral role in maintaining glucose homeostasis and preventing hypoglycemia. Pancreatic A cells also participate in the local regulation of islet activities as glucagon acts through glucagon receptors on A, B and D type cells within the islets. Recent studies have also shown that Pancreatic A cells play a role in the generation and regeneration of B type cells. Following beta cell injuries pancreatic A cells increase in numbers and produce Glucagon-like peptide-1 (GLP-1), which increases the proliferation and cytoprotection of beta cells. In response to extreme injury of B type cells pancreatic A cells can transform (transdifferentiate) into functioning B type cells. Until recently, glucagon has been considered a pancreas-specific hormone; however, extrapancreatic glucagon has been reported in patients who had undergone complete, and glucagon-positive cells been identified in the human stomach, indicating that Type A enteroendocrine cells are not restricted to the pancreas.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002067") -AnnotationAssertion(Annotation( "DOI:10.3389/fphys.2013.00102") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK572070") Annotation( "https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3164530") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/cardiac-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.3389/fphys.2013.00102") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK572070") Annotation( "https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3164530") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/cardiac-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Cardiac myocytes, also known as cardiac muscle cells, form the majority of the cardiac tissue and are responsible for the contractile function of the heart. These myocytes are columnar-shaped cells with centrally located nuclei, and they exhibit visibly striated cytoplasm due to the organized array of myofibrils, which are composed of filaments of actin and myosin. Cardiac myocytes connect with each other through specialized structures called intercalated discs, facilitating electrical and mechanical continuity and enabling synchronous contraction of the heart. They also possess a high number of mitochondria to meet their high energy demand for continuous heartbeat action. Cardiac myocytes are capable of automaticity, meaning they have the unique ability to spontaneously and rhythmically generate their electrical impulses, a characteristic led by pacemaker cells. The electrical signals initiated and propagated in these cells are responsible for heartbeats. They also respond to the electrical signals transmitted by the autonomic nervous system and chemical signals like hormones. Thus, cardiac myocytes not only participate in maintaining the heart’s function but also play a role in modulating heart rate and strength of contraction in response to the body's changing needs. Consequently, malfunction of these cells leads to serious cardiac diseases such as heart failure and arrhythmias.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002098") -AnnotationAssertion(Annotation( "DOI:10.1016/j.bbamcr.2015.11.025") Annotation( "DOI:10.1111/pace.14107") Annotation( "DOI:10.1172/JCI25417") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/cardiac-muscle") Annotation( "https://www.sciencedirect.com/topics/neuroscience/cardiac-action-potential") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.bbamcr.2015.11.025") Annotation( "DOI:10.1111/pace.14107") Annotation( "DOI:10.1172/JCI25417") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/cardiac-muscle") Annotation( "https://www.sciencedirect.com/topics/neuroscience/cardiac-action-potential") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Regular atrial cardiac myocytes, also known as atrial myocytes, are specialized muscle cells found in the atria – the upper chambers of the heart. They contract and relax during the heart's cycle, modulating the pumping of blood through the atria and into the ventricles. Additionally, endowed with inherent rhythmic electrical activity, atrial myocytes contribute significantly to the initiation and propagation of the heart’s electrical impulses. A distinguishing feature that separates atrial myocytes from other cardiac myocytes is their ability to synthesize and secrete atrial natriuretic peptide (ANP) in response to atrial stretch or dilatation. ANP acts as a potent vasodilator and diuretic, helping to maintain blood pressure and volume homeostasis. The phenomena of atrial stretch or distension, which triggers ANP release, is often in response to excess blood volume entering the heart, providing a mechanism at the cellular level which actively regulates systemic cardiovascular balance. Like all cardiac myocytes, atrial myocytes are embedded in a dense network of connective tissue that provides structural support. These cells are characterized by a single, centrally located nucleus, and overall, have a rod-like appearance with branching ends that connect with adjacent cells to form a continuous, synchronized ensemble. Their cytoplasm is abundant with mitochondria, reflecting the high energy demand associated with constant contraction and relaxation. At the ultrastructural level, atrial myocytes display striations due to the regular arrangement of actin and myosin proteins, which facilitate the contraction process essential for the heart's pump function.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002129") -AnnotationAssertion(Annotation( "DOI:10.1038/s41569-022-00770-1") Annotation( "DOI:10.1186/s12872-015-0124-z") Annotation( "DOI:10.3389/fphys.2022.863265/full") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/vascular-endothelial-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41569-022-00770-1") Annotation( "DOI:10.1186/s12872-015-0124-z") Annotation( "DOI:10.3389/fphys.2022.863265/full") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/vascular-endothelial-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial cells of the vascular tree, commonly referred to as vascular endothelial cells, line the entire circulatory system, from the heart to the smallest capillaries. These cells have a unique, flattened shape, and are tightly bound together, forming a thin layer known as the endothelium. The endothelium is responsible for maintaining the interior surface of blood vessels, and plays a critical role in ensuring the smooth flow of blood. Vascular endothelial cells act as a semi-permeable membrane, controlling the passage of materials and the transit of white blood cells into and out of the bloodstream. These cells are also involved in blood coagulation. When a blood vessel is damaged, endothelial cells promote clotting to prevent excessive bleeding and facilitate wound healing. Other functions include regulation of blood pressure and blood volume by releasing vasodilators and vasoconstrictors to either widen or constrict blood vessels, accordingly. Furthermore, the endothelial cells are crucial in the formation of new blood vessels, a process known as angiogenesis. This is particularly important in wound healing and the growth of new tissues during development or after injury. In the context of diseases such as cancer, angiogenesis helps facilitate tumor growth by providing nutrients and oxygen.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002139") -AnnotationAssertion(Annotation( "DOI:10.1101/cshperspect.a028241") Annotation( "DOI:10.1146/annurev-physiol-021014-071931") Annotation( "DOI:10.1152/ajplung.00329.2019") Annotation( "DOI:10.1159/000196486") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1101/cshperspect.a028241") Annotation( "DOI:10.1146/annurev-physiol-021014-071931") Annotation( "DOI:10.1152/ajplung.00329.2019") Annotation( "DOI:10.1159/000196486") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Ciliated columnar cells of the tracheobronchial tree compose the inner lining of the tracheobronchial tree, the system of airways consisting of trachea, bronchi and bronchioles that allow passage of air into the lungs, where gas exchange occurs. A defining feature of these endo-epithelial cells are the tiny hair-like structures covering their surface, known as cilia. The nucleus is located at the base of the cell, and the area above it is rich in mitochondria and well-developed endoplasmic reticulum, both crucial for the energy-intensive process of cilia operation. The primary function of these ciliated cells is to keep the respiratory tract clean via mucociliary clearance or the respiratory escalator. These cells create a two-layered 'coat': The lower \"sol\" layer is watery where the cilia can beat in coordinated waves, and the upper \"gel\" layer is thick and sticky, trapping inhaled particles, such as dust, bacteria, viruses, and other potentially harmful substances. This rhythmical beating of the cilia then moves the mucus and trapped particles upwards and out of the respiratory tract, which is then either coughed out or swallowed. Damage or dysfunction of these ciliated cells, as seen in diseases such as primary ciliary dyskinesia, cystic fibrosis, or chronic bronchitis, may lead to reduced or ineffective mucociliary clearance and an increased susceptibility to respiratory infections.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002145") -AnnotationAssertion(Annotation( "DOI:10.1038/leu.2010.214") Annotation( "DOI:10.1126/science.75570") Annotation( "DOI:10.1182/blood-2004-06-2480") Annotation( "DOI:10.1182/blood-2009-08-239194") Annotation( "https://www.sciencedirect.com/topics/engineering/endosteum") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/leu.2010.214") Annotation( "DOI:10.1126/science.75570") Annotation( "DOI:10.1182/blood-2004-06-2480") Annotation( "DOI:10.1182/blood-2009-08-239194") Annotation( "https://www.sciencedirect.com/topics/engineering/endosteum") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endosteal cells, also known as osteogenic cells, are a specialized type of cell that resides in the endosteum of the bone marrow cavity. The endosteum is a thin vascular membrane of connective tissue that lines the inner surface of the bony tissue that forms the medullary cavity of long bones. This specific location assigns endosteal cells critical roles in the bone maintenance and regeneration process as they actively participate in bone remodeling, a process that involves both the formation and resorption of bone tissue. Endosteal cells function by regulating the activity of both osteoblasts and osteoclasts. Osteoblasts are cells that deposit new bone, and osteoclasts are responsible for bone resorption, a process important for the maintenance, repair, and remodelling of bones. When new bone tissue is required, such as in instances of bone fractures or increased mechanical stress, endosteal cells differentiate into osteoblasts to facilitate the bone formation process. Conversely, when bone resorption is necessary, these cells are known to release signals that lead to the recruitment and activation of osteoclasts. In addition to this, endosteal cells play a significant role in hematopoiesis, which is the formation of blood cellular components. They function in this process by providing a niche for hematopoietic stem cells, serving to support their maintenance and differentiation. Understanding the functionality of endosteal cells is important in the field of regenerative medicine and stem cell therapy, especially in diseases affecting bone remodeling and hematopoiesis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002157") -AnnotationAssertion(Annotation( "DOI:10.1007/978-3-211-99390-3_83") Annotation( "DOI:10.1038/nature16161") Annotation( "DOI:10.1111/j.1469-7580.2005.00403.x") Annotation( "DOI:10.3389/fphys.2015.00087") Annotation( "DOI:10.5114/ceji.2022.124416") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/978-3-211-99390-3_83") Annotation( "DOI:10.1038/nature16161") Annotation( "DOI:10.1111/j.1469-7580.2005.00403.x") Annotation( "DOI:10.3389/fphys.2015.00087") Annotation( "DOI:10.5114/ceji.2022.124416") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Brush cells, also referred to as tuft cells or multivesicular cells, are a specialized type of epithelial cell mainly noted for their characteristic 'brush border' composed of microvilli. These cells reside in the epithelial lining of tissue organs such as the respiratory tract, gastrointestinal tract, and the bile ducts. The name derives from their distinct appearance under the microscope, which resembles a brush due to the dense layer of microvilli protruding into the lumen. A key function of brush cells is chemosensation: They express a variety of signaling molecules and receptors that enable them to detect specific chemical stimuli in the environment and act as sensory transducers. Many of these receptors are responsive to luminal content, which makes brush cells vital for regulating and coordinating appropriate physiological responses to changes in these substances. Recent research has also elucidated an important role for these cells in immunity. Intestinal brush cells have been found to be the source of a cytokine called interleukin-25, which can initiate type 2 immune responses during parasitic infections. This immune function, along with the chemosensing abilities, signifies that brush cells could serve crucial roles in health and disease, although much research is still required to fully elucidate their myriad roles in physiology and pathology. Overall, brush cells are a versatile cell type, whose distinct morphology and functional capabilities allow them to perform a unique set of functions within the body.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002204") -AnnotationAssertion(Annotation( "DOI:10.1016/j.biocel.2007.10.034") Annotation( "DOI:10.1111/aos.14600") Annotation( "DOI:10.1155/2021/9951032") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.biocel.2007.10.034") Annotation( "DOI:10.1111/aos.14600") Annotation( "DOI:10.1155/2021/9951032") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The anterior lens cells, also known as lens epithelial cells, are an integral part of the eye's structure and play a critical role in the organ's physiological functioning. They are situated in the anterior portion of the lens, precisely in the lens capsule, and stretch over the lens' frontal surface. The primary function of anterior lens cells is to facilitate eye accommodation by controlling the shape and thickness of the eye lens. The cells are involved in continuous proliferation, migration, and differentiation into lens fiber cells, helping to maintain lens growth and transparency. Anterior lens cells have a prolonged life span and reveal an extraordinary ability to function indefinitely, a characteristic that is critical for maintaining lens transparency. Damage or disturbance to the anterior lens cells can give rise to lens opacity, eventually leading to cataracts, which is a common cause of vision loss.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002223") -AnnotationAssertion(Annotation( "DOI:10.1016/B978-1-4377-1926-0.10005-0") Annotation( "DOI:10.1016/j.biocel.2007.10.034") Annotation( "DOI:10.1098/rstb.2010.0324") Annotation( "DOI:10.1136/bmjophth-2020-000459") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/lens-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/B978-1-4377-1926-0.10005-0") Annotation( "DOI:10.1016/j.biocel.2007.10.034") Annotation( "DOI:10.1098/rstb.2010.0324") Annotation( "DOI:10.1136/bmjophth-2020-000459") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/lens-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Secondary lens fibers are a specialized type of elongated cells located within the structure of the eye's lens. They play an essential role in vision by enabling light transmission and focus on the retina, which allows for clear, distinct vision. These cells are characterized by their lack of nuclei and organelles, their orderly alignment, and their high protein content, particularly crystallins. Lens fibers are differentiated from equatorial epithelial cells of the lens in a process that involves cell elongation, denucleation and tight packing, which serve to reduce light scattering occurrences in the eye. This differentiation accelerates substantially after birth in comparison to during embryogenesis and continues throughout life, contributing to the growth of the eye lens. Secondary lens fibers are specifically those cells that are newly differentiated and cover the old lens fiber core. This process also leads to the removal of light-obstructing cellular components, mainly nuclei and organelles, making these fibers transparent and ideal for their function. The fibers carry out their primary function of light refraction via a high concentration of specialized proteins known as crystallins, which form a gradient of refractive index together with the cytoplasm. This index manipulation facilitates precise light focus onto the retina. Secondary lens fibers also contribute to the lens's shape and biomechanical properties through intercellular interactions, particularly at the sutures where the ends of the fibers meet. Any disruption to secondary lens fibers can lead to vision problems, including cataracts, which is the clouding of the lens resulting from the aggregation of crystallin proteins.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002225") -AnnotationAssertion(Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532252/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK537180/") Annotation( "https://www.sciencedirect.com/topics/neuroscience/iris-dilator-muscle") Annotation( "https://www.sciencedirect.com/topics/neuroscience/iris-sphincter-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532252/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK537180/") Annotation( "https://www.sciencedirect.com/topics/neuroscience/iris-dilator-muscle") Annotation( "https://www.sciencedirect.com/topics/neuroscience/iris-sphincter-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The smooth muscle cell of sphincter of pupil is a specialized cell type typically found within the eye, more specifically within the sphincter muscle of the iris. These cells can constrict and dilate in response to light signals, tightly regulating the amount of light reaching the retina. Contraction narrows the pupil in bright conditions (pupillary constriction), protecting the retina from excessive light. In low light, relaxation allows dilation (pupillary dilation), enhancing visual perception in dim conditions. These cells are also innervated by parasympathetic fibers from the oculomotor nerve. Light signals to the Edinger-Westphal nucleus stimulate the release of acetylcholine, contracting the cells. On the contrary, sympathetic stimulation induces relaxation. This precise control plays a crucial role in optimizing visual function, highlighting their essential role in vision biology.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002243") -AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2013.07.004") Annotation( "DOI:10.1038/s41575-018-0081-y") Annotation( "DOI:10.1038/s41580-020-0278-0") Annotation( "DOI:10.1101/gad.1674008") Annotation( "DOI:10.1111/j.1365-2184.2009.00642.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2013.07.004") Annotation( "DOI:10.1038/s41575-018-0081-y") Annotation( "DOI:10.1038/s41580-020-0278-0") Annotation( "DOI:10.1101/gad.1674008") Annotation( "DOI:10.1111/j.1365-2184.2009.00642.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Intestinal crypt stem cells, also known as crypt base columnar cells, are a unique type of cell, characterized by the highly specific marker LGR5, found in the intestinal epithelium. Situated at the bottom of the minute pockets known as crypts of Lieberkühn, these are undifferentiated cells that have the ability to perpetually self-renew, as well as differentiate into various other cell types that constitute the epithelial lining of the intestine. The fundamental role of intestinal crypt stem cells is to provide a constant supply of new cells to maintain the cellular turnover of the intestinal epithelium, a tissue known for rapid self-renewal. These stem cells are nurtured and protected by specialized epithelial and mesenchymal cells, and together constitute the intestinal stem cell niche. An important function of intestinal crypt stem cells is to sustain the balance between cell division and programmed cell death, called apoptosis, to ensure the integrity of the intestinal lining. Given their active proliferation rate, these stem cells initiate the creation of diverse differentiated cell types, including enterocytes (the primary absorptive cells in the intestinal lumen), goblet cells (that produce mucus to protect the epithelial layer), enteroendocrine cells (involved in producing gastrointestinal hormones), and Paneth cells (involved in secreting antimicrobial peptides). This diversity in output regulates the physiological activities of the gut ranging from nutrient absorption, hormone secretion, bacterial balance, to immunity. Research studies suggest that dysregulation in intestinal crypt stem cell proliferation and differentiation is associated with several intestinal disorders including intestinal cancer, and various enteropathies.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002250") -AnnotationAssertion(Annotation( "DOI:10.1007/s10456-021-09780-y") Annotation( "DOI:10.1016/j.jhep.2016.07.009") Annotation( "DOI:10.1038/s41575-018-0020-y") Annotation( "DOI:10.1038/s41598-020-57652-0") Annotation( "DOI:10.1186/1476-5926-1-1") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s10456-021-09780-y") Annotation( "DOI:10.1016/j.jhep.2016.07.009") Annotation( "DOI:10.1038/s41575-018-0020-y") Annotation( "DOI:10.1038/s41598-020-57652-0") Annotation( "DOI:10.1186/1476-5926-1-1") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial cells of sinusoids, often referred to as sinusoidal endothelial cells (SECs), are a specialized type of endothelial cell that primarily reside in the capillaries of the liver, spleen, and bone marrow. These cells form the innermost monolayer of the sinusoidal blood vessels, also known as sinusoids, that mediate the exchange of nutrients, metabolites, and waste materials between the blood and the surrounding organ tissue. SECs possess distinctive morphological features including a large, flattened shape, an abundance of fenestrations or pores, and the lack of a continuous basement membrane. These structural characteristics distinguish sinusoidal endothelial cells from other, more regularly structured endothelial cells in the body. Due to their special fenestrated structure, SECs facilitate the exchange of macromolecules, like lipoproteins and hyaluronan, between plasma and the surrounding organ parenchymal cells. These fenestrations act like sieves, allowing the passage of substances based on their size and charge. It's noteworthy that the permeability characteristics of SECs can be regulated dynamically according to the body's homeostatic needs. Beyond their key role in exchange mechanisms, endothelial cells of sinusoids are involved in several other functions. In the liver, for example, they help in the removal and endocytic degradation of waste macromolecules, immune response, and regulation of liver regeneration. They also participate in the formation and remodeling of blood vessels, a process known as angiogenesis. Additionally, in recent years, it has been discovered that these cells may have a role in disease conditions like cirrhosis and liver cancer.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002262") -AnnotationAssertion(Annotation( "DOI:10.1016/B978-0-12-819402-7.00001-2") Annotation( "DOI:10.1016/B978-012369442-3/50154-9") Annotation( "DOI:10.1016/j.mce.2015.06.028") Annotation( "DOI:10.1038/s42255-019-0148-2") Annotation( "DOI:10.3389/fendo.2023.1192311") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/B978-0-12-819402-7.00001-2") Annotation( "DOI:10.1016/B978-012369442-3/50154-9") Annotation( "DOI:10.1016/j.mce.2015.06.028") Annotation( "DOI:10.1038/s42255-019-0148-2") Annotation( "DOI:10.3389/fendo.2023.1192311") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Pancreatic polypeptide (PP) cells (also called F or gamma cells) are unique endocrine cells located within the Islets of Langerhans in the pancreas. PP cells are one of the rarer pancreatic cell types and are more prevalent in the head and neck of the pancreas. They are critical in normal pancreatic physiological functions and are involved in the development of pancreatic endocrine disorders. The primary function of PP cells is the production and secretion of the pancreatic polypeptide hormone (PP). This hormone plays a crucial role in several gastrointestinal functions and metabolic responses. The release of the PP hormone is stimulated after eating, especially in protein-rich meals, leading to it being present in large amounts during digestion. The fundamental role of PP is to self-regulate pancreatic secretion activities ensuring its exocrine and endocrine functions are under control. The pancreatic polypeptide hormone from PP cells also aids in adapting to low physical activity and fasting by reducing the production of insulin and glucagon and inhibiting the hepatic glucose production. Additionally, this hormone influences gut motility by slowing down the gastric emptying and reducing small intestinal transit, thereby controlling the pace at which nutritional substances are absorbed. Because of these functions, any malfunction or irregularity in PP cells can result in various disorders such as diabetes and pancreatic diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002275") -AnnotationAssertion(Annotation( "DOI:10.1016/S1569-2590(05)10005-6") Annotation( "DOI:10.1111/j.1444-0938.2002.tb02384.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S1569-2590(05)10005-6") Annotation( "DOI:10.1111/j.1444-0938.2002.tb02384.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Pigmented ciliary epithelial cells, which are a type of pigment cell, have a crucial function within the eye's ciliary body. The ciliary body, located behind the iris, is one of the eye’s most vital structures and consists of two types of epithelial cells: the pigmented and the non-pigmented ciliary epithelial cells. The pigmented ciliary epithelial cells form the outer layer of the ciliary body and exhibit a black or brown pigmentation due to the melanin they contain. One key function of pigmented ciliary epithelial cells is to aid in the formation of aqueous humor, an intraocular fluid that nourishes the cornea and lens, and maintains intraocular pressure which is essential for the eye’s shape and light refraction. These cells facilitate this function in conjunction with the non-pigmented ciliary epithelial cells. Together, the pigmented and non-pigmented ciliary epithelial cells form a bilayer epithelium that allows the secretion of aqueous humor through a bi-directional fluid transport mechanism. Pigmented ciliary epithelial cells also contribute to the blood-aqueous barrier, a physiological frontier that controls the entry and exit of various substances from the blood to the aqueous humor and vice versa. The pigmentation in these cells, intensified by melanin, helps to absorb scattered light coming into the eye, reducing any potential damage and glare.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002303") -AnnotationAssertion(Annotation( "DOI:10.1002/iub.1404") Annotation( "DOI:10.1093/molehr/gan048") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK9906/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/iub.1404") Annotation( "DOI:10.1093/molehr/gan048") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK9906/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Embryonic cells in metazoans, which include all multicellular animals, are biological units in their earliest developmental stages from conception until the organism is fully formed. They are derived from a fertilized egg, which undergoes a series of cell divisions to produce a multicellular organism. During the early phases of embryonic development, these cells exhibit pluripotency, meaning they possess the potential to differentiate into a diverse array of specialized cell types that constitute different body tissues and structures. The balance between cell proliferation and differentiation in embryonic cells is crucial for successful development. When cells continue to divide without differentiating, overgrowth may occur leading to conditions such as teratomas (tumors of embryonic cells). Conversely, if cell differentiation proceeds at the expense of cell proliferation, developmental anomalies could occur regarding size or mass of the organism or its parts.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002321") -AnnotationAssertion(Annotation( "DOI:10.1165/rcmb.2013-0049OC") Annotation( "DOI:10.1186/s12931-014-0160-8") Annotation( "DOI:10.2353/ajpath.2010.090870") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1165/rcmb.2013-0049OC") Annotation( "DOI:10.1186/s12931-014-0160-8") Annotation( "DOI:10.2353/ajpath.2010.090870") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The basal epithelial cells of the tracheobronchial tree are critical components found in the lining of the airway passages, including the trachea and bronchi. They are keratin-5-positive, nonciliated, cuboidal cells and typically tightly attached to the basement membrane. In humans, the proportion of basal cells in the respiratory epithelium gradually decreases going down the tracheobronchial tree: they represent approximately 34% of the cells in the trachea, 27% in the large airways, and 10% in the small airways, although it is worth noting that there are differences in the compositions of the tracheobronchial epithelia between different species. Basal epithelial cells serve as the basal layer of the tracheobronchial epithelium, providing both structural and regenerative support for the epithelial tissue that lines the upper regions of the respiratory tract. They serve as progenitor or stem cells that are capable of differentiating into multiple cell types, such as ciliated and secretory cells. This gives them a central role in homeostatic maintenance of the epithelium, and in repairing damaged epithelium after an injury or during disease. This regenerative capacity is crucial in maintaining the integrity of the tracheobronchial surface, especially given its continual exposure to inhaled irritants and microorganisms. In addition to their primary restorative function, basal epithelial cells are also involved in the initial immune response within the tracheobronchial tree. Equipped with pattern recognition receptors, these cells can identify and respond to pathogenic organisms, triggering an immune response and producing a range of inflammatory mediators, such as cytokines and chemokines, to help combat infections.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002329") -AnnotationAssertion(Annotation( "DOI:10.1016/j.medici.2017.08.003") Annotation( "DOI:10.1016/j.ydbio.2009.06.033") Annotation( "DOI:10.1101/cshperspect.a036723") Annotation( "DOI:10.1161/ATVBAHA.121.313788") Annotation( "DOI:10.1161/CIRCRESAHA.117.312136") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.medici.2017.08.003") Annotation( "DOI:10.1016/j.ydbio.2009.06.033") Annotation( "DOI:10.1101/cshperspect.a036723") Annotation( "DOI:10.1161/ATVBAHA.121.313788") Annotation( "DOI:10.1161/CIRCRESAHA.117.312136") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endocardial cells, often referred to as endothelial cells of the heart, constitute the innermost lining layer of the heart tissues, forming the endocardium. They play a critical role in maintaining heart functionality and homeostasis. These cells, flat and squamous in structure, are adjoined closely to form a tight barrier that separates the heart's muscular layer, the myocardium, from the blood flowing through the heart chambers. Functionally, endocardial cells are key players in several crucial physiological processes within the heart. Synthesizing matrix molecules that contribute to the structural formation of the heart, these cells actively partake in maintaining its structural integrity. They also exhibit unique metabolic activity, which aids in maintaining optimal cardiac functioning in both healthy and pathological conditions. Importantly, they are known for their involvement in modulating myocardial contraction and relaxation, that is crucial for normal heart rhythm and function. They achieve this by producing factors like nitric oxide (NO) which, amongst other things, aids in the regulation of blood pressure, prevents blood clotting and inhibits the adherence of blood cells to the vessel wall. In pathological states, endocardial cells can undergo a transformation process known as endothelial-mesenchymal transition (EndMT), which is inherently involved in several cardiac diseases. In this transition, they acquire the ability to migrate and differentiate into several other types of cells contributing to disease progression. Moreover, their dysfunction can lead to endocarditis, an inflammation of the endocardium, as well as increase the risk of other heart diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002350") -AnnotationAssertion(Annotation( "DOI:10.1016/bs.ai.2014.09.003") Annotation( "DOI:10.1038/nri3667") Annotation( "DOI:10.1111/sji.13094") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/bs.ai.2014.09.003") Annotation( "DOI:10.1038/nri3667") Annotation( "DOI:10.1111/sji.13094") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Cortical thymic epithelial cells (cTECs) are a vital cell type located in the thymus, a lymphoid organ that plays a key role in the development of T cells, which are essential for the adaptive immune system. cTECs also have a role in forming barriers and lining surfaces. In the thymus, their presence contributes to the distinctive architecture of both the cortex and medulla. cTECs also play a significant part in the selection and development of T cells. During T cell maturation, these cells express both self and non-self proteins via the major histocompatibility complex (MHC) to immature T cells. cTECs then stimulate the T cells that can recognize such proteins, an important event known as positive selection. This process aids in the creation of a diverse T cell receptor (TCR) repertoire that can react to a wide range of antigens, therefore ensuring effective immunity. Apart from aiding in T cells' positive selection, cTECs also contribute to eliminating self-reactive T cells, a role essential in preventing autoimmune diseases. These cells induce an apoptosis-driven process known as \"negative selection\" which eliminates T cells that are too highly reactive to self-antigens.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002364") -AnnotationAssertion(Annotation( "DOI:10.1007/s11427-013-4482-4") Annotation( "DOI:10.4049/jimmunol.2100692") Annotation( "DOI:10.7554/eLife.60188") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s11427-013-4482-4") Annotation( "DOI:10.4049/jimmunol.2100692") Annotation( "DOI:10.7554/eLife.60188") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Medullary thymic epithelial cells (mTECs) are distinct cells situated in the thymic medulla, a vital organ where T cells mature. mTECs are recognized by their unique appearance, characterized by large, plump cells with abundant cytoplasm and irregularly shaped nuclei. They inhabit a microenvironment closely interacting with various thymic cell types, including conventional dendritic cells and thymocytes. The primary function of mTECs revolves around negative selection of developing T cells, thus preventing autoimmune responses. mTECs allow developing T cells to recognize and be responsive to foreign antigens while remaining tolerant to the body's own tissues. They achieve this through a process called promiscuous gene expression where they express and display a vast array of self-antigens. This process is regulated by a transcriptional regulator called autoimmune regulator (Aire), which contributes to immunological self-tolerance. mTECs themselves have a self-renewing ability and sustain the continuous export of mature T cells. They also express a number of genes that encode for chemokines and other signaling molecules which draw in and retain developing T cells for the process of negative selection. The three-dimensional network formed by these cells provides a physical platform for such selection. Together, mTECs hold a critical place in the maintenance of immune homeostasis, demonstrating the conceptual links that exist between population of stromal cells, thymocytes development, tissue-restricted antigen expression, and negative selection.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002365") -AnnotationAssertion(Annotation( "DOI:10.1073/pnas.070447210") Annotation( "DOI:10.1093/humupd/dmr031") Annotation( "DOI:10.1177/1535370220938741") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/uterine-contraction") Annotation( "https://www.sciencedirect.com/topics/veterinary-science-and-veterinary-medicine/myometrium") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1073/pnas.070447210") Annotation( "DOI:10.1093/humupd/dmr031") Annotation( "DOI:10.1177/1535370220938741") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/uterine-contraction") Annotation( "https://www.sciencedirect.com/topics/veterinary-science-and-veterinary-medicine/myometrium") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Myometrial cells are specialized smooth muscle cells located in the myometrium, the middle layer of the uterine wall. Unique to female reproductive physiology, they play a key role in pregnancy and childbirth. These cells are distinctive in their capability to substantially increase in size and number during pregnancy, preparing the uterus to accommodate the growing fetus. As pregnancy progresses, myometrial cells demonstrate a progressive growth in uterine mass through cellular hypertrophy. They are also responsible for the production of extracellular matrix proteins like collagen, which aids in supporting cell structure and function, further facilitating uterine enlargement. During childbirth, myometrial cells are responsible for the expansion and contraction of the uterus. They can propagate action potentials and generate considerable force, which is critical to their function in the reproductive system. After pregnancy, these cells can reduce in size, a process known as uterine involution. Given the important role these cells play in female reproductive physiology, dysregulation of the myometrial cell function can contribute to uterine pathologies such as uterine fibroids.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002366") -AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1111/febs.15731") Annotation( "DOI:10.1165/ajrcmb.25.5.f218") Annotation( "DOI:10.2147/COPD.S38938") Annotation( "DOI:10.3109/01902148.2013.791733") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1111/febs.15731") Annotation( "DOI:10.1165/ajrcmb.25.5.f218") Annotation( "DOI:10.2147/COPD.S38938") Annotation( "DOI:10.3109/01902148.2013.791733") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The respiratory goblet cell is a highly specialized cell type found primarily within the respiratory tract, including the nose, trachea, and lungs. Named because their shape resembles a goblet - a drinking vessel with a wide body and narrow neck - these cells form a vital part of the respiratory system's protective mechanisms. They are found within the columnar epithelium lining these organs, which forms a barrier between the internal body and the exterior environment. A principal function of the respiratory goblet cell is the production and secretion of mucus, a carbohydrate-rich, viscous and gel-like substance that plays a critical role in trapping dust, bacteria, viruses, and other airborne particles that are inhaled. The mucus secreted by the goblet cells covers the lining of the respiratory tract, effectively catching these particles and preventing them from reaching the lungs and causing infection. Mucus also provides hydration and lubrication to the respiratory tract surfaces, which is important in maintaining the tissue's health and functioning. In addition to their mucus-secreting capabilities, respiratory goblet cells also play a significant role in the body's inflammatory responses. When the respiratory system is exposed to irritants or pathogens, the number and activity of goblet cells often increase, leading to a higher production of mucus. This is a protective response designed to trap and neutralize the harmful substances more effectively. However, in conditions like chronic obstructive pulmonary disease (COPD) and asthma, an overproliferation of goblet cells, also known as goblet cell hyperplasia, can lead to excessive mucus production and airway obstruction.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002370") -AnnotationAssertion(Annotation( "DOI:10.1038/nrm3980") Annotation( "https://www.genome.gov/genetics-glossary/Somatic-Cells") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK557896/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nrm3980") Annotation( "https://www.genome.gov/genetics-glossary/Somatic-Cells") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK557896/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Somatic cells represent the majority of the cell types in the human body. They are fundamental building blocks of organs, tissues, and other bodily structures, with every organ being composed of distinct subpopulations of these cells. The primary function of somatic cells is to maintain the function and survival of an organism. They carry significant information in the form of DNA, and through the process of mitosis, contribute to the repair and regeneration of body tissues. Further, some types of somatic cells work in a collaborative manner to form complex functional structures such as the skin and lining of the gut, demonstrating a higher level of organization. However, it's important to note that somatic cells are distinct from germ cells, which are responsible for sexual reproduction by forming sperm or eggs. Any alterations in the DNA of somatic cells, due to mutations, will not affect the offspring as they aren't involved in transmission of genetic information to the next generation. While most somatic cells contain two copies of each chromosome (diploid), a certain subset may possess a single set of chromosomes (haploid), specifically found in male ants, bees, and other hymenopterans. Hence, diversity is a defining characteristic of somatic cells, reflecting in their structures, roles, and genetic makeup.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002371") -AnnotationAssertion(Annotation( "DOI:10.1038/nrgastro.2012.168") Annotation( "DOI:10.1111/j.1365-2982.2012.01986.x") Annotation( "DOI:10.1371/journal.pcbi.1009644") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nrgastro.2012.168") Annotation( "DOI:10.1111/j.1365-2982.2012.01986.x") Annotation( "DOI:10.1371/journal.pcbi.1009644") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Enteric smooth muscle cells are a specialized type of cell found in the gastrointestinal tract. They are an integral part of the enteric nervous system, which regulates the functions of the gastrointestinal system. These cells have a unique structural organization that enables their primary function: the contraction and relaxation necessary for the propulsion of gastrointestinal contents. Smooth muscle cells in the enteric system possess unique attributes that distinguish them from other smooth muscle cells in the body. One crucial function is peristalsis, where these cells contract in a coordinated manner, generating a wave of contraction and relaxation. This peristaltic movement facilitates the movement of food particles through the digestive tract, playing a vital role in the digestion and absorption processes in animals. Additionally, these cells are responsible for maintaining the tone of gastrointestinal sphincters, controlling passage and preventing one-sided movement of the contents. Enteric smooth muscle cells are also involved in local immune responses of the gastrointestinal tract. They can communicate with local immune cells and secrete cytokines or other signaling molecules in response to infectious organisms, which makes them crucial mediocellular players in gastrointestinal homeostasis and response to disease. Their interactions with other cell types, such as neurons, myofibroblasts, and interstitial cells of Cajal, further contribute to these cells' role in maintaining the physiological functions of the gastrointestinal system. Overall, enteric smooth muscle cells provide an indispensable contribution to the complex operations of the digestive tract.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002504") -AnnotationAssertion(Annotation( "DOI:10.1002/hep.31252") Annotation( "DOI:10.1016/j.ajpath.2023.02.012") Annotation( "DOI:10.1016/j.biocel.2010.06.020") Annotation( "DOI:10.1016/j.gastha.2022.07.015") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/hep.31252") Annotation( "DOI:10.1016/j.ajpath.2023.02.012") Annotation( "DOI:10.1016/j.biocel.2010.06.020") Annotation( "DOI:10.1016/j.gastha.2022.07.015") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Intrahepatic cholangiocytes represent a subset of the biliary epithelial cells that form a network of tubes in the liver called the biliary tree The intrahepatic cholangiocytes reside specifically in the intrahepatic bile ducts and play a crucial role in liver physiology and bile production and secretion, thereby aiding in the digestion and absorption of fats in the small intestine. These specific cholangiocytes arise from bipotent hepatoblasts, whereas extrahepatic cholangiocytes share an embryologic origin with the ventral pancreas. Recent research suggests that there is also heterogeneity within populations of intrahepatic cholangiocytes with different transcriptional profiles, proliferative capacity, and biological function; for example, subpopulations differ in calcium-mobilizing receptors. The primary function of intrahepatic cholangiocytes is related to bile formation and maintenance of its flow. In addition to their secretory and absorptive activities, they are involved in the regulation of bile composition, volume, and alkalinization, contributing to the neutralization of the acidic chyme (partly digested food) that enters the intestine from the stomach. Intrahepatic cholangiocytes are also recognized for their role in liver regeneration and repair, often proliferating in response to injury. Intrahepatic cholangiocytes take part in certain pathological conditions, like primary biliary cirrhosis and cholangiocarcinoma. Changes in these cells often lead to abnormalities in bile formation and transport, causing cholestatic liver diseases. Overall, the primary and secondary functional activities of intrahepatic cholangiocytes are vital in maintaining liver function, digestive processes, and contributing to the body's response to liver injury.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002538") -AnnotationAssertion(Annotation( "DOI:10.1016/j.devcel.2005.05.017") Annotation( "DOI:10.1111/febs.12414") Annotation( "DOI:10.7150/ijbs.49871") Annotation( "https://training.seer.cancer.gov/anatomy/cells_tissues_membranes/tissues/muscle.html") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/aortic-smooth-muscle-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.devcel.2005.05.017") Annotation( "DOI:10.1111/febs.12414") Annotation( "DOI:10.7150/ijbs.49871") Annotation( "https://training.seer.cancer.gov/anatomy/cells_tissues_membranes/tissues/muscle.html") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/aortic-smooth-muscle-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Aortic smooth muscle cells are specialized and highly differentiated muscle cells that are located in the tunica media layer of the aorta. They consist of spindle-shaped cells with a centrally located nucleus. These cells are rich in actin and myosin, muscle contractile proteins, that allow them to exert force and change shape. Aortic smooth muscle cells play a vital role in maintaining blood pressure and circulation, and are essential for vascular integrity and function. The primary function of these cells is contraction and relaxation, which enables the regulation of blood flow and pressure in the aorta, and helps in the distribution of oxygen and nutrients throughout the body. Additionally, a critical characteristic of smooth muscle cells is their plasticity. They can undergo phenotypic modulation in response to changes in their environment or vascular injury, switching from a contractile to a synthetic phenotype. The contractile phenotype is characterized by high contractility and low proliferation rate, while the synthetic phenotype is marked by increased cell proliferation and matrix synthesis but reduced contractile function. Aortic smooth muscle cells have a significant role in the physiological and pathological processes of the cardiovascular system. In the normal physiological state, they contribute to the elasticity and flexibility of the aorta. Conversely, in pathological states, changes in the function and structure of aortic smooth muscle cells are connected with various vascular diseases, including atherosclerosis, hypertension, and aneurysm.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002539") -AnnotationAssertion(Annotation( "DOI:10.1007/s00441-008-0706-5") Annotation( "DOI:10.1038/s41569-022-00770-1") Annotation( "DOI:10.1038/s41598-021-01360-w") Annotation( "DOI:10.3390/ijms20184411") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00441-008-0706-5") Annotation( "DOI:10.1038/s41569-022-00770-1") Annotation( "DOI:10.1038/s41598-021-01360-w") Annotation( "DOI:10.3390/ijms20184411") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Vein endothelial cells form a thin layer of squamous cells, the endothelium, lining the interior surface of veins throughout the body. They shape the inner cellular lining of the entire vascular system, including the heart, playing a crucial role in blood circulation. The unique characteristic compact arrangement of these cells enables veins to act as a barrier between the blood (that can contain foreign substances) and the surrounding venous tissue and maintain the integrity of the vascular system. The primary function of vein endothelial cells is to control the exchange of substances between the bloodstream and the surrounding tissues. They facilitate selective transportation of molecules depending on their size and solubility, including gases, nutrients, hormones, and waste products. Vein endothelial cells also play a protective role by inhibiting the translocation of toxins or pathogens from blood to tissues. Additionally, these cells are involved in blood coagulation and inflammation response, primarily by producing substances that inhibit blood clot formation under normal conditions and initiating clotting when necessary. Another significant function of vein endothelial cells is the regulation of blood flow and blood pressure. They produce and release several substances, including nitric oxide and prostacyclin, which help in controlling vasodilation and vasoconstriction, thereby regulating blood pressure. These cells are also responsible for angiogenesis, the formation of new blood vessels, which is crucial during wound healing and in the formation of granulation tissue. Consequently, any dysfunction in vein endothelial cells can lead to severe health problems like atherosclerosis, hypertension, and thrombosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002543") -AnnotationAssertion(Annotation( "DOI:10.1016/j.biopha.2019.108765") Annotation( "DOI:10.1016/j.tibtech.2006.01.010") Annotation( "DOI:10.1038/s41536-019-0083-6") Annotation( "DOI:10.1096/fj.202100332R") Annotation( "DOI:10.1111/dgd.12049") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.biopha.2019.108765") Annotation( "DOI:10.1016/j.tibtech.2006.01.010") Annotation( "DOI:10.1038/s41536-019-0083-6") Annotation( "DOI:10.1096/fj.202100332R") Annotation( "DOI:10.1111/dgd.12049") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Mesenchymal stem cells of adipose tissue, also known commonly as adipose-derived stem cells (ADSCs), are a population of adult stem cells that can be obtained easily from adipose tissues. They have many of the same regenerative properties as other mesenchymal stem cells, but are more easily accessible than bone marrow-derived stem cells. Adipose-derived stem cells show immense promise in the field of regenerative medicine due to their ability to differentiate into adipocytes, chondrocytes, myocytes, osteoblasts, and other cell types. ADSCs also possess immunomodulatory and homeostatic functions: they have the ability to suppress immune responses and provide a therapeutic environment for tissue repair and regeneration, as well as supporting the proliferation of adipocytes and the overall expansion of adipose tissue and contributing to the tissue's ability to react to demands of energy storage and mobilization. Dysfunction of these cells may contribute to metabolic complications observed in obesity and diabetes. ADSCs have been used for therapeutic applications such as pathological wound healing, severe refractory acute graft-versus-host disease, and idiopathic thrombocytopenic purpura.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002570") -AnnotationAssertion(Annotation( "DOI:10.1002/glia.23892") Annotation( "DOI:10.1016/B978-0-444-52902-2.00005-9") Annotation( "DOI:10.1016/j.biocel.2006.05.007") Annotation( "DOI:10.1186/1742-2094-8-110") Annotation( "DOI:10.1186/s13064-020-00140-y") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/glia.23892") Annotation( "DOI:10.1016/B978-0-444-52902-2.00005-9") Annotation( "DOI:10.1016/j.biocel.2006.05.007") Annotation( "DOI:10.1186/1742-2094-8-110") Annotation( "DOI:10.1186/s13064-020-00140-y") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Schwann cells, also known as neurolemmocytes, are a type of glial cell located in the peripheral nervous system. These cells play a significant role in the healthy functioning of nerves by producing myelin, a fatty substance that forms a coating around nerve fibers. Myelin serves as an insulator and enhances the speed and efficiency of electrical nerve impulses; a single Schwann cell can myelinate a single axon. Myelination starts by the elongation and envelopment of the Schwann cell around the axon, followed by the synthesis and deposition of myelin layers. Some studies suggest that Schwann cells may regulate neuronal action potential, muscular contraction, and the sensitive response. While Schwann cells are most commonly known for the formation of the myelin sheath, some Schwann cells do not form myelin: Remak Schwann cells, a class of nonmyelinating Schwann cells, ensheath axons with smaller diameter, such as C fiber nociceptors in sciatic nerves and form Remak bundles. Schwann cells are found along both motor and sensory neurons and are crucial for the advancement and recovery of peripheral nerve injuries, due to their capacity to support nerve regeneration. In cases of nerve injury, Schwann cells play a vital role in recovery by initiating Wallerian degeneration, a process in which the part of the axon distal to the injury site degrades and is then cleared away. Following this, Schwann cells can guide the regrowth of the nerve, providing a conducive environment for axon regeneration. They remodel themselves into a regenerative phenotype, proliferate, and organize themselves into bands of Büngner that provide physical and chemical guidance for the regrowths of axons. This function of Schwann cells in the repair and regeneration of nervous system highlights their therapeutic potential in peripheral nerve injury treatments.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002573") -AnnotationAssertion(Annotation( "DOI:0.1016/j.preteyeres.2015.08.001") Annotation( "DOI:10.1016/j.preteyeres.2012.08.004") Annotation( "DOI:10.1038/nature04482") Annotation( "DOI:10.5301/EJO.2010.6049") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:0.1016/j.preteyeres.2015.08.001") Annotation( "DOI:10.1016/j.preteyeres.2012.08.004") Annotation( "DOI:10.1038/nature04482") Annotation( "DOI:10.5301/EJO.2010.6049") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Retinal blood vessel endothelial cells constitute the innermost lining of the blood vessels found in the retina, the light-sensitive layer of tissue at the back of the eye. The primary role of these cells revolves around their location within the retinal vascular system, forming the structure of the blood-retinal barrier, a subset of the larger blood-ocular barrier system. The endothelial cells are particularly involved in the growth of new retinal blood vessels from pre-existing ones (angiogenesis); this is critical in managing the amount of oxygen and essential nutrients delivered to the ocular tissues. They are crucial for maintaining homeostasis in the retinal environment by controlling the exchange of molecules between the blood and the retina. Furthermore, they mediate immune cell trafficking, supporting the immune privilege of the eye by preventing the unrestricted infiltration of inflammatory cells into the retina, thus maintaining ocular health and normal vision. The malfunction or dysfunction of retinal blood vessel endothelial cells is implicated in numerous ocular pathologies, particularly diabetic retinopathy, which is one of the leading causes of blindness worldwide. In such conditions, the compromise in the integrity of the blood-retinal barrier and excess angiogenesis can lead to retinal edema and pathological neovascularization, resulting in vision loss.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002585") -AnnotationAssertion(Annotation( "DOI:10.1111/febs.16018") Annotation( "DOI:10.1152/physrev.00021.2004") Annotation( "DOI:10.3389/fphar.2021.727870/full") Annotation( "https://www.sciencedirect.com/topics/chemistry/retinal-pigment") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1111/febs.16018") Annotation( "DOI:10.1152/physrev.00021.2004") Annotation( "DOI:10.3389/fphar.2021.727870/full") Annotation( "https://www.sciencedirect.com/topics/chemistry/retinal-pigment") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Retinal pigment epithelial (RPE) cells form a single layer of cells at the back of the eye sandwiched between the neurosensory retina and the choroid, playing a significant role in maintaining vision health. These pigment-laden cells are highly specialized and perform an array of metabolic and transport functions essential for the maintenance of the photoreceptor cells (rods and cones) in the retina. The pigmentation of RPE cells actively aids in the absorption of excess light and the prevention of light scattering, thus enhancing the eye's optical properties. The retinal pigment epithelium forms a key part of the blood/retina barrier. The cells have long sheet-like microvilli on their apical membrane that project into the light-sensitive outer segments of the photoreceptors, forming a close structural interaction. The basolateral membrane of the RPE interacts with the underlying Bruch’s membrane, which separates the RPE cells from fenestrated endothelium of the choriocapillaris. RPE cells support the photoreceptor by providing them with oxygen and nutrients (such as glucose, retinol and fatty acids) and removing waste products. They also recycle the visual pigment, in a process called the \"visual cycle\", where the RPE cells play a vital role in the regeneration of visual pigment (11-cis retinol) following the absorption of light. This is essential for the maintenance of photoreceptor excitability. Beyond this, RPE cells take part in the phagocytosis process, where they digest the shed ends of photoreceptor outer segments, thus, preventing the build-up of waste residue that could otherwise harm retinal health. They also secrete various factors, including growth factors required to maintain the structural integrity of choriocapillaris endothelium and photoreceptors, as well as immunosuppressive factors that play an important role in establishing the immune privilege of the eye.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002586") -AnnotationAssertion(Annotation( "DOI:10.1016/j.apsb.2012.12.007") Annotation( "DOI:10.1124/pr.115.010652") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/pulmonary-artery-smooth-muscle-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.apsb.2012.12.007") Annotation( "DOI:10.1124/pr.115.010652") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/pulmonary-artery-smooth-muscle-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Smooth muscle cells of the pulmonary artery form a principal component of the pulmonary artery's media layer, the middle section of the artery wall typically found between endothelial inner layer (intima) and the outermost connective tissue layer (adventitia). The unique structure of these cells, defined by fusiform shape and the absence of striations, sets them apart from other muscle cell types, including cardiac and skeletal. The primary function of smooth muscle cells of the pulmonary artery is to regulate the diameter of the pulmonary artery, which in turn controls the flow of blood into the lungs. They accomplish this task through the contraction and relaxation, indicative of vasoconstriction and vasodilation respectively. When these cells contract, the diameter of the artery narrows, leading to reduced blood flow. Conversely, relaxation of these cells widens the artery, facilitating increased blood flow. Such blood flow modulation is crucial for the maintenance of blood pressure and ensuring that the lungs receive an adequate blood supply for efficient oxygen exchange. As a result, abnormal function or proliferation of these cells can contribute to conditions such as pulmonary hypertension, a condition characterized by high blood pressure in the arteries leading to the lungs. This can occur due to hypoxia (lack of enough oxygen), leading to excessive constriction or proliferation of the smooth muscle cells.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002591") -AnnotationAssertion(Annotation( "DOI:10.1152/japplphysiol.00313.2006") Annotation( "DOI:10.1183/09031936.00019810") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK537353/") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/bronchial-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/japplphysiol.00313.2006") Annotation( "DOI:10.1183/09031936.00019810") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK537353/") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/bronchial-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Bronchial smooth muscle cells are specialized types of cells found in the bronchi of the respiratory system in mammals. These bronchi are tubular structures that function to carry air from the trachea into the lungs. As part of the smooth muscle tissue, bronchial smooth muscle cells are involuntary cells, meaning they function with minimal direct voluntary control. They are characterized by their elongated spindle-shaped structure, with each cell containing a single central nucleus. The primary function of bronchial smooth muscle cells is to control airway diameter, which directly influences the flow of air into and out of the lungs. Muscular contractions and relaxations mediated by these cells enable the bronchi to constrict or expand, facilitating the regulation of respiratory airflow. Additionally, these cells play a pivotal role in immune responses as they produce and release various substances that can instigate or lessen inflammation of airway tissue. Furthermore, bronchial smooth muscle cells can proliferate and lay down collagen and other extracellular matrix proteins, contributing to the structural integrity of the bronchial tubes. In pathological conditions, such as asthma and chronic obstructive pulmonary disease, the function and biology of bronchial smooth muscle cells change. Asthma, for example, is associated with an abnormal increase in the mass of the bronchial smooth muscles, leading to narrowing of the airways and resultant breathing difficulties. These cells also exhibit altered contractile behavior and an augmented immune response during inflammatory lung diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002598") -AnnotationAssertion(Annotation( "DOI:10.1016/s1357-2725(02)00259-5") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK9961") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/airway-smooth-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/s1357-2725(02)00259-5") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK9961") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/airway-smooth-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Smooth muscle cells of the trachea have a characteristic 'smooth' (non-striated) appearance under the microscope, a distinguishing feature that differentiates them from skeletal and cardiac muscle cells. The smooth appearance is due to the organization of actin and myosin filaments in the cell, which allows for a controlled, contractile function. These cells play a key role in controlling the diameter of the trachea, thus regulating the airflow. They are responsible for the involuntary constriction and relaxation of the trachea influencing the air passage during various physiological behaviors like breathing, coughing, and sneezing. This autonomic control allows for adaptations to specific body requirements without conscious control, such as increasing air flow during exercise or restricting it during rest. In conditions such as asthma, chronic bronchitis, and emphysema, there are changes in smooth muscle cells. For example, the mass of smooth muscle cells increases in chronic airway diseases, possibly in response to chronic inflammation. This increase may indicate a pathological condition or be a part of the body's response to injury and repair.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002600") -AnnotationAssertion(Annotation( "DOI:10.1177/0036850419850431") Annotation( "DOI:10.1203/00006450-199811000-00001") Annotation( "DOI:10.3791/56639") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532927/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1177/0036850419850431") Annotation( "DOI:10.1203/00006450-199811000-00001") Annotation( "DOI:10.3791/56639") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532927/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Uterine smooth muscle cells are specialized cells that make up the majority of the uterus and in particular, the myometrium. The myometrium is a thick, involuntary muscle layer, principally consisting of verticillate cell bundles that sculpt the helical architecture of the uterus. This dense muscular tissue, packed with uterine smooth muscle cells, is responsible for the significant contractile force witnessed during the physical processes of labor and childbirth. In the non-pregnant uterus, they exist in a quiescent phase. Upon implantation, these cells are primarily responsible for the expansion consequently allowing the uterus to accommodate the growing fetus. Towards the end of pregnancy, under the influence of hormonal and mechanical signals, uterine smooth muscle cells transition from a relaxed phase to an active contractile phase, culminating in the delivery of the fetus. At the cellular level, these transitions involve significant changes in the electrophysiological properties of uterine smooth muscle cells. Governed by a complex interplay of ion channels, transporters, and signaling proteins, these cells adjust their electrical activity, conduction properties, and contracting mechanisms. Any issues with these processes can result in problems such as preterm birth or labor dysfunction.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002601") -AnnotationAssertion(Annotation( "DOI:10.1002/cyto.a.20952") Annotation( "DOI:10.1007/s12015-006-0015-x") Annotation( "DOI:10.1172/JCI107470") Annotation( "DOI:10.3390/app10030938") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK53254/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cyto.a.20952") Annotation( "DOI:10.1007/s12015-006-0015-x") Annotation( "DOI:10.1172/JCI107470") Annotation( "DOI:10.3390/app10030938") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK53254/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial cells of the umbilical vein form the inner lining of the veins found in the umbilical cord. They are involved in tube formation and migration which are essential for angiogenesis, the process of generating new blood vessels; this is critical during the fetal stage for the development of the circulatory system. Endothelial cells of the umbilical vein also play a role in controlling the passage of white blood cells into tissues during inflammatory responses. Because human umbilical vein endothelial cells (HUVECs) can easily be derived from the umbilical cord, and because they express common endothelial cell markers, they have been used as an epithelial cell model for studies on cell proliferation, migration, angiogenesis, and inflammation. They have been used as models for vascular diseases like atherosclerosis, for investigating how tumor cells infiltrate blood vessels and form metastases. Thus, while endothelial cells of the umbilical vein play a critical role in vascular biology, they also represent a powerful tool in disease studies and potential treatment strategies.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002618") -AnnotationAssertion(Annotation( "DOI:10.1111/j.1745-7254.2007.00625.x") Annotation( "DOI:10.1111/jnc.15689") Annotation( "DOI:10.3389/fimmu.2022.997786") Annotation( "DOI:10.4103/1673-5374.226386") Annotation( "DOI:10.4103/1673-5374.322423") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1111/j.1745-7254.2007.00625.x") Annotation( "DOI:10.1111/jnc.15689") Annotation( "DOI:10.3389/fimmu.2022.997786") Annotation( "DOI:10.4103/1673-5374.226386") Annotation( "DOI:10.4103/1673-5374.322423") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Mature microglial cells represented one of the macrophage populations within the central nervous system (CNS). Microglial cells serve as the primary immune defense mechanism within the CNS, playing active roles in maintaining brain homeostasis and responding to pathological processes. In response to pathogens and injuries, microglial cells mature and change their morphology from a ramified shape with small processes (normal state) to an amoeboid shape (mature state). Mature microglial cells possess multiple functional abilities, thereby enabling them to execute diverse roles in order to maintain the health of surrounding neural cells. They act to clear away cellular debris, dead neurons, or plaque formations through the process of phagocytosis. They are also continuously scanning their environment and are able to detect subtle changes in the neural landscape caused by injury, disease, or infection. Upon detection of such changes, these cells become activated and present antigens and secrete cytokines and chemokines to initiate an inflammation response, which sustains the brain’s defense against injurious agents. Their role is not solely destructive or defensive: mature microglial cells also contribute to neurodevelopment by guiding the formation of neural circuits, synaptogenesis, and remodeling. Furthermore, they assist in managing synaptic plasticity, a key process in learning and memory functions. While the dysregulation of microglial cells has been implicated in various neurodegenerative diseases, they are also regarded as potential therapeutic targets for such conditions, given their crucial roles in maintaining CNS health.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002629") -AnnotationAssertion(Annotation( "DOI:10.1186/s12931-022-02042-5") Annotation( "DOI:10.3389/falgy.2021.787128") Annotation( "DOI:110.1038/s41385-020-00370-7") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1186/s12931-022-02042-5") Annotation( "DOI:10.3389/falgy.2021.787128") Annotation( "DOI:110.1038/s41385-020-00370-7") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Respiratory basal cells are highly specialized cells that serve a crucial role in the human airway epithelium, predominantly found lining the surfaces of the trachea and bronchi. These cells exhibit a distinctive columnar shape and are attached directly to the basal lamina. They are characterized by the presence of high amounts of cytoplasmic keratins, predominantly keratin 5 and 14, and transcription factor tumor protein (tp63), which set them apart from other types of lung epithelial cells such as ciliated cells and secretory cells. Basal cells are essential for maintaining airway integrity. They make up one third of all respiratory epithelial cells and serve as stem cells as they can transform into different cell types, like goblet cells, ciliated cells, and club cells, when needed for homeostatic maintenance of the epithelial barrier or to repair and restore a healthy cellular environment after injury. Their position at the interface between the internal environment and the external atmosphere makes these cells a first line of defense against air-borne irritants, allergens, and pathogens. Their robust, intrinsic reparative properties facilitate effective recovery from such adversities. However, damage to respiratory basal cells or abnormalities in their function are associated with several respiratory pathologies such as asthma, chronic obstructive pulmonary disease, and lung cancer. Therefore, understanding these cells' function, and their role in disease, is crucial for the development of new therapeutic strategies for respiratory disorders.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002633") -AnnotationAssertion(Annotation( "DOI:10.1007/s12079-019-00511-z") Annotation( "DOI:10.1016/j.cmet.2013.08.001") Annotation( "DOI:10.1101/cshperspect.a006569") Annotation( "DOI:10.3389/fcell.2021.642352") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s12079-019-00511-z") Annotation( "DOI:10.1016/j.cmet.2013.08.001") Annotation( "DOI:10.1101/cshperspect.a006569") Annotation( "DOI:10.3389/fcell.2021.642352") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial stalk cells are specialized vascular cells vital in angiogenesis, a process through which new blood vessels are formed from pre-existing ones; specifically, endothelial stalk cells are involved in sprouting angiogenesis, where they help form the body of new blood vessels. Vascular sprouting relies on the coordinated activity of migrating endothelial tip cells at the forefront and proliferating stalk cells that elongate the sprout. The process is tightly controlled by different growths factors: Vascular Endothelial Growth Factor acts on endothelial cells, inducing them to become endothelial tip cells that initiate sprouting. After sprouting initiation, activation of Notch signaling suppresses differentiation toward a tip cell phenotype and some of the endothelial cells differentiate into stalk cells, which follow tip cells, multiply, and elongate to provide a structural backbone to the growing vessel sprout. In contrast to endothelial tip cells, which migrate and lead the angiogenic sprout, endothelial stalk cells behind the sprouts continue forming the tube or lumen for blood flow and facilitate maturation and stability of the new vessel. The delicate balance between the activities of stalk and tip cells during angiogenesis is crucial to build a functional vascular network. Dysfunctions in endothelial stalk cells can lead to pathological conditions such as impaired wound healing, unregulated tumor growth, and metastasis due to abnormal angiogenesis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002671") -AnnotationAssertion(Annotation( "DOI:10.1002/ca.21011") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK507782/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK554405/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/ca.21011") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK507782/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK554405/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Tongue muscle cells are responsible for essential functions like mastication, deglutition (swallowing), phonation (speech), and taste receptivity. They can be classified into two functional classes - extrinsic and intrinsic muscle cells. The extrinsic tongue muscle cells originate outside the tongue and are primarily involved in determining the position of the tongue within the mouth. Muscle groups such as the genioglossus, hyoglossus, and styloglossus are composed of these extrinsic muscle cells and grant the tongue its wide range of motion. The intrinsic tongue muscle cells, on the other hand, originate and function within the tongue itself, influencing the shape of the tongue during speech, eating, and swallowing. These cells can contract to alter the tongue's shape, making it long, short, curled, or flat, as needed. Together, these two types of muscle cells allow the tongue to perform its vital functions effectively. Moreover, they play a critical role in maintaining oral health by facilitating physical cleaning of the oral cavity and assisting in the mechanical breakdown of food.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0002673") -AnnotationAssertion(Annotation( "DOI:10.1016/j.jcf.2019.09.010") Annotation( "DOI:10.1038/s41586-018-0393-7") Annotation( "DOI:10.1038/s41598-023-30603-1") Annotation( "DOI:10.1146/annurev-pathol-042420-094031") Annotation( "DOI:10.3389/fmars.2020.00709") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.jcf.2019.09.010") Annotation( "DOI:10.1038/s41586-018-0393-7") Annotation( "DOI:10.1038/s41598-023-30603-1") Annotation( "DOI:10.1146/annurev-pathol-042420-094031") Annotation( "DOI:10.3389/fmars.2020.00709") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Ionocytes are specialized cells predominantly found in the mammalian respiratory and renal systems as well as in the gills, skin, and intestinal tract of fish. These cells play crucial roles in maintaining ion and acid-base homeostasis. Ionocytes demonstrate remarkable plasticity and are able to adapt themselves in response to changes in environmental conditions such as pH, salinity, ion concentration, and temperature. These cells work by selectively absorbing specific ions from the environment, thus maintaining the body's internal ionic balance. The most commonly absorbed ions include sodium (Na+), chloride (Cl-), calcium (Ca2+), and hydrogen (H+). Moreover, ionocytes contribute to acid-base regulation. In response to acidosis or alkalosis, ionocytes can either excrete or retain hydrogen (H+) and bicarbonate (HCO3-) ions to readjust the blood pH. Further, experimental evidence suggests a functional complexity of ionocytes, implying diverse roles beyond ion regulation. Recent research highlights ionocytes' involvement in ammonia excretion and the regulation of extracellular fluid volume, highlighting their contribution to the overall homeostatic process. Malfunctioning ionocytes have been implicated in various diseases, including cystic fibrosis which is caused by mutations in the chloride channel CFTR, an ionocyte marker.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005006") -AnnotationAssertion(Annotation( "DOI:10.1016/j.krcp.2013.07.005") Annotation( "DOI:10.2215/CJN.05760513") Annotation( "DOI:10.2215/CJN.08580813") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.krcp.2013.07.005") Annotation( "DOI:10.2215/CJN.05760513") Annotation( "DOI:10.2215/CJN.08580813") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Renal principal cells are the major cell type in the initial collecting tubule and the cortical and outer medullary collecting ducts, as well as the connecting tubule, in the nephrons of the kidneys. A key function of renal principal cells is the regulation of water balance in the body. These cells express aquaporin-2 (AQP2) water channels, which facilitate the reabsorption of water from the fluid within the renal tubule lumen back into the blood. The translocation of AQP2 to the cell membrane is controlled by the hormone vasopressin: In response to high vasopressin levels, AQP2 moves to the cell membrane, allowing water to pass through effectively and be reabsorbed, therefore concentrating the urine. Conversely, in low vasopressin conditions, AQP2 is removed from the membrane, reducing water reabsorption and diluting the urine. Renal principal cells are also involved in the regulation of sodium and potassium levels in the body. They have sodium channels (ENaC) in their luminal membranes which permit the reabsorption of sodium ions from the tubule fluid back into the blood, resulting in a net reabsorption of sodium ions into the systemic circulation. This process is controlled, in part, by the hormone aldosterone. Additionally, renal principal cells contain potassium channels that facilitate the secretion of potassium ions into the urine. The activities of these channels, in coordination with other cell types in the nephron, are critical for maintaining electrolyte balance and overall body homeostasis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005009") -AnnotationAssertion(Annotation( "DOI:10.1152/ajprenal.2000.279.1.F195") Annotation( "DOI:10.1161/HYPERTENSIONAHA.121.16492") Annotation( "DOI:10.1681/ASN.V1011") Annotation( "DOI:10.2215/CJN.08880914") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/ajprenal.2000.279.1.F195") Annotation( "DOI:10.1161/HYPERTENSIONAHA.121.16492") Annotation( "DOI:10.1681/ASN.V1011") Annotation( "DOI:10.2215/CJN.08880914") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Renal intercalated cells are specialized cells located in the collecting duct system of the kidneys. The primary role of intercalated cells is to reabsorb bicarbonate ions and secrete hydrogen ions, thereby maintaining the acid-base homeostasis in the blood. They comprise two main subtypes primarily distinguished by their functional and morphological attributes: alpha and beta intercalated cells. Alpha intercalated cells are more predominant when the body is in a state of acidosis, a condition characterized by an increased acidity of the blood. These cells are specialized in secreting excessive hydrogen ions into the urine through a mechanism involving vacuolar H+-ATPase and H+/K+-ATPase pumps on their apical membranes. They simultaneously reabsorb bicarbonate ions from the tubular fluid and return them to the bloodstream via mechanisms involving carbonic anhydrase II and bicarbonate/chloride exchangers on the basolateral membrane. This dual process helps to increase blood pH towards normal levels. When the body is in a state of alkalosis, a condition characterized by lowered levels of hydrogen ions in the blood, beta intercalated cells are more predominant. They primarily reabsorb hydrogen ions from the tubular fluid through vacuolar H+-ATPase and H+/K+-ATPase pumps on their basolateral membranes, while secreting bicarbonate ions into the urine via pendrin, a bicarbonate/chloride exchanger in the apical membrane. These mechanisms work together to decrease blood pH towards normal levels. In summary, renal intercalated cells play a critical role in the delicate balance of the body’s pH, safeguarding the body from potential harm caused by acidemia or alkalemia.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005010") -AnnotationAssertion(Annotation( "DOI:10.1016/j.semnephrol.2019.04.005") Annotation( "DOI:10.1152/physiol.00008.2011") Annotation( "DOI:10.1172/JCI63492") Annotation( "DOI:10.3390/diseases2020071") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.semnephrol.2019.04.005") Annotation( "DOI:10.1152/physiol.00008.2011") Annotation( "DOI:10.1172/JCI63492") Annotation( "DOI:10.3390/diseases2020071") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Renal alpha-intercalated cells are located within the connecting tubules and collecting ducts of the kidneys, which are components of the kidney's complex nephron system. Along with beta-intercalated cells, they play a critical role in the body’s acid-base balance. Renal alpha-intercalated cells contain an abundance of proton pumps and enzymes like carbonic anhydrase, which aid in the transport of hydrogen ions for secretion in the urine. The bicarbonate/chloride transporters on their apical membrane meanwhile take up bicarbonate ions from the urine and supply them back into the blood. Through this mechanism, these cells contribute significantly to the neutrality of blood pH, and dysfunction of renal alpha-intercalated cells often leads to distal renal tubular acidosis, a condition resulting in acidic blood and alkaline urine.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005011") -AnnotationAssertion(Annotation( "DOI:10.1016/j.cub.2014.08.047") Annotation( "DOI:10.1038/nrm.2017.21") Annotation( "DOI:10.1101/cshperspect.a028233") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.cub.2014.08.047") Annotation( "DOI:10.1038/nrm.2017.21") Annotation( "DOI:10.1101/cshperspect.a028233") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The multi-ciliated epithelial cell are terminally differentiated epithelia that line brain ventricles, the respiratory tract and parts of the female and male reproductive organs in animals, playing critical roles in the maintenance of homeostasis through ciliary motion. They are characterized by the presence of hundreds of motile cilia, hair-like microtubule-based organelles that beat in a coordinated fashion to direct fluid flow over the cell surface. Multi-ciliated epithelial cells have fundamental roles in the proper functioning of many organ systems. In the respiratory system, they line the airways and orchestrate the coordinated movement of mucus, effectively clearing the airways of inhaled particles and pathogens. These cells are also vital in the ventricular system of the brain where they facilitate cerebrospinal fluid circulation, thus contributing to the maintenance of the brain's microenvironment. In the fallopian tube, multi-ciliated epithelial cells aid in the transport of oocytes from the ovary to the uterus, a process crucial to reproduction. The formation and function of multi-ciliated epithelial cells is a highly regulated process that involves several stages of development including cell specification, centriole multiplication, and ciliogenesis. Any disruption in these processes can result in dysfunctional or decreased numbers of cilia, which can lead to a myriad of health issues, ranging from chronic respiratory infections to infertility.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005012") -AnnotationAssertion(Annotation( "DOI:10.1007/s00125-008-1238-y") Annotation( "DOI:10.1152/physrev.00012.2004") Annotation( "DOI:10.1210/en.2018-00833") Annotation( "DOI:10.3389/fendo.2022.904004") Annotation( "DOI:10.3390/ijms20081867") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00125-008-1238-y") Annotation( "DOI:10.1152/physrev.00012.2004") Annotation( "DOI:10.1210/en.2018-00833") Annotation( "DOI:10.3389/fendo.2022.904004") Annotation( "DOI:10.3390/ijms20081867") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Pancreatic epsilon cells are a specialized type of endocrine cell found in the islets of Langerhans, a region of the pancreas responsible for hormone production. These clusters of cells constitute only about 1% of the pancreatic islet cell population, making them a relatively small yet significant component of the pancreas. Pancreatic epsilon cells have a round or ovoid shape with occasional cytoplasmic extensions and are characterized by small and spherical granules. The principal function of pancreatic epsilon cells involves the synthesis and release of the hormone ghrelin, a peptide hormone predominantly produced in the stomach; pancreatic epsilon cells are one of the few sites outside the gastrointestinal tract known to produce this hormone. Ghrelin has multiple vital roles, playing a significant part in generating hunger sensations, promoting fat storage, and influencing various metabolic processes. It also stimulates the release of Growth Hormone (GH) from the anterior pituitary gland. During fetal development, when they form a layer around the islet, epsilon cells are an important source of ghrelin, likely secreting the hormone into the circulation; their numbers decrease in adults. While research on pancreatic epsilon cells is still ongoing, these cells have been implicated in several disease states, most notably Type 2 diabetes and metabolic syndrome. The dysfunction or reduction in the number of pancreatic epsilon cells can lead to anomalies in ghrelin production, impacting overall metabolic homeostasis and glucose regulation.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005019") -AnnotationAssertion(Annotation( "DOI:10.1016/j.stem.2014.04.010") Annotation( "DOI:10.1093/jb/mvr001") Annotation( "DOI:10.1242/dev.031369") Annotation( "DOI:10.1242/dev.114215") Annotation( "DOI:10.5966/sctm.2015-0051") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.stem.2014.04.010") Annotation( "DOI:10.1093/jb/mvr001") Annotation( "DOI:10.1242/dev.031369") Annotation( "DOI:10.1242/dev.114215") Annotation( "DOI:10.5966/sctm.2015-0051") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Hepatoblasts are immature precursor cells that predominate during the early stages of liver development, specifically in the embryonic phase of life. They first arise from the endoderm, one of the three primary germ layers in the very early embryo, and then differentiate into two distinct mature liver cell types - the hepatocytes and cholangiocytes. During liver organogenesis hepatoblasts proliferate and migrate into the septum transversum to form the liver bud. Proliferation and differentiation of these cells are regulated by several soluble factors, such as hepatocyte growth factor, which is a mitogen of both hepatoblasts and mature hepatocytes. As they start to differentiate into hepatocytes and cholangiocytes, the cells begin to express hepatic markers like albumin and alpha-fetoprotein. Although hepatoblasts are specified embryonic liver cells that are bipotential for hepatocytes and cholangiocytes, a subset of liver cells (called oval cells) has been identified in adults that express stem cell markers, such as CD133 and cKIT, and has been suggested to have the same potential as hepatoblasts to differentiate into hepatocytes and cholangiocytes.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0005026") -AnnotationAssertion(Annotation( "DOI:10.1038/srep32884") Annotation( "DOI:10.1096/fba.2020-00058") Annotation( "DOI:10.1210/endo.141.9.7634") Annotation( "DOI:10.12659/MSMBR.901142") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/srep32884") Annotation( "DOI:10.1096/fba.2020-00058") Annotation( "DOI:10.1210/endo.141.9.7634") Annotation( "DOI:10.12659/MSMBR.901142") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Preosteoblast is a critical cell type involved in bone formation, crucial in a process known as ossification or osteogenesis. They are derived from mesenchymal stem cells and are an intermediate stage marking the transition from an osteoprogenitor cell to a fully differentiated osteoblast. The presence of preosteoblasts signifies the site of new bone deposition, highlighting their crucial role in skeletal development and in response to bone injury repairs. The primary function of preosteoblasts is to differentiate into osteoblasts, which are responsible for secreting osteoid (the unmineralized portion of bone matrix) and regulating the process of bone mineralization. Preosteoblasts are regulated by various growth factors and hormones, including bone morphogenic proteins (BMPs), fibroblast growth factors (FGFs), parathyroid hormone (PTH) and Vitamin D. However, the function of preosteoblasts is not limited to bone formation alone. In addition to osteogenesis, preosteoblasts significantly contribute to the maintenance of bone homeostasis through regulating the activity of osteoclasts - the cells responsible for bone resorption. This regulation is mediated through RANK/RANKL/OPG pathway signaling. Osteoprotegerin (OPG) released from the preosteoblasts acts as a decoy receptor for RANKL impairing osteoclast formation, hence preventing excessive bone resorption. This regulatory role indicates the dual functionality of preosteoblasts, which are indispensable for both the formation of new bone and the maintenance of existing bone tissue.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0007010") -AnnotationAssertion(Annotation( "DOI:10.1007/s12522-011-0102-9") Annotation( "DOI:10.1016/j.it.2017.01.009") Annotation( "DOI:10.1016/j.preghy.2010.10.003") Annotation( "DOI:10.1080/19336918.2015.1089376") Annotation( "DOI:10.1080/19336918.2015.1120397") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s12522-011-0102-9") Annotation( "DOI:10.1016/j.it.2017.01.009") Annotation( "DOI:10.1016/j.preghy.2010.10.003") Annotation( "DOI:10.1080/19336918.2015.1089376") Annotation( "DOI:10.1080/19336918.2015.1120397") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Extravillous trophoblasts (EVTs), a specialized subset of trophoblast cells, play crucial roles in establishing and sustaining pregnancy. Firstly, they anchor the fetus to the maternal tissue, providing structural stability. Secondly, they are instrumental in modifying the maternal spiral arteries to ensure an adequate supply of maternal blood to the placenta and the developing fetus. EVTs invade maternal decidua and myometrium and replace the endothelial lining of the spiral arteries, transforming these high-resistance, narrow vessels into low-resistance, wide vessels, favouring higher blood flow. Thirdly, these cells also play an immunological role, creating an environment conducive to the acceptance of the semi-allogeneic fetus, by expressing non-classical MHC molecules, thereby averting any potential maternal immune response towards fetal tissues. While EVTs are commonly associated with healthy pregnancies, abnormalities in their function or development have been connected to problematic pregnancies. Conditions like pre-eclampsia and fetal growth restriction (FGR) may arise if EVT cells do not effectively invade the maternal decidua or adequately remodel.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0008036") -AnnotationAssertion(Annotation( "DOI:10.1016/j.coph.2013.09.008") Annotation( "DOI:10.1038/mi.2017.73") Annotation( "DOI:10.1038/nature24489") Annotation( "DOI:10.1111/j.1365-2613.2011.00767.x") Annotation( "DOI:10.1146/annurev-physiol-021115-105439") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.coph.2013.09.008") Annotation( "DOI:10.1038/mi.2017.73") Annotation( "DOI:10.1038/nature24489") Annotation( "DOI:10.1111/j.1365-2613.2011.00767.x") Annotation( "DOI:10.1146/annurev-physiol-021115-105439") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The enteroendocrine cell of the small intestine is a specialized type of cell that forms part of the lining of the intestinal wall. These cells are interspersed among absorptive cells, mucus-secreting goblet cells, and other gut cell types, forming the crypt-villus axis, which is the functional unit of the small intestinal epithelium. Functionally, enteroendocrine cells play a pivotal role in the gut-endocrine system, which is responsible for the digestive process. These cells translate the luminal nutrient status into hormonal signals thereby acting as chemosensors. They contain secretory granules at their base which release hormones into the bloodstream following chemo-sensation. The hormones they secrete perform diverse functions including regulating gastric secretion, gut motility, insulin release, appetite control and other local and systemic effects. In addition to their endocrine functions, enteroendocrine cells also play a part in the modulation of the immune response in the gut. They can secrete cytokines and chemokines that have a role in directing the immune response to invading pathogens. Moreover, emerging research suggests that these cells play a role in maintaining the balance between gut resident bacteria, known as the gut microbiota, and the host. As such, a comprehensive understanding of the enteroendocrine cells of the small intestine underpins knowledge in nutrition, endocrinology, and various gastroenterological conditions.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009006") -AnnotationAssertion(Annotation( "DOI:10.1002/ibd.20197") Annotation( "DOI:10.1038/nature09637") Annotation( "DOI:10.1038/nrmicro2546") Annotation( "DOI:10.1093/ecco-jcc/jjac190.0884") Annotation( "DOI:10.1136/gut.48.2.176") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/ibd.20197") Annotation( "DOI:10.1038/nature09637") Annotation( "DOI:10.1038/nrmicro2546") Annotation( "DOI:10.1093/ecco-jcc/jjac190.0884") Annotation( "DOI:10.1136/gut.48.2.176") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Paneth cells of the colon are specialized secretory epithelial cells that can be found in the large intestine, albeit in small numbers compared with the small intestine where Paneth cells are more abundant. Under disease conditions such as inflammatory bowel disease, Paneth cells increase in numbers in the colon. They reside in the crypts of Lieberkühn, which are glandular structures embedded within the mucosal layer of the small and large intestine. Paneth cells play a crucial role in maintaining intestinal homeostasis and acting as a frontline of defense in our bodies against ingested microbes. They achieve this through the secretion of antimicrobial peptides and proteins such as lysozyme and defensins, which can directly kill or inhibit the growth of various bacteria, fungi, and viruses. They degranulate these substances into the intestinal lumen, especially under conditions of pathogenic invasion or damage to the epithelial lining. Moreover, Paneth cells are essential for the maintenance of intestinal stem cells, which continually replace the epithelium of the intestine. They create a niche for these stem cells at the base of the crypts, secreting various signaling molecules and factors that are essential for the growth and differentiation of intestinal stem cells. In essence, the existence and function of Paneth cells are fundamental to the overall gut health and immune defense system, by providing both antimicrobial functions and a conducive environment for the renewal of the intestinal lining. The role of Paneth cells specifically in the colon is of interest in the context of inflammatory bowel disease, such as ulcerative colitis and Crohn’s disease: Their presence in the colon is thought to represent Paneth cell metaplasia and indicates a chronic inflammatory state.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009009") -AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2014.02.057") Annotation( "DOI:10.1053/j.gastro.2018.08.016") Annotation( "DOI:10.1172/jci.insight.150894") Annotation( "DOI:10.3389/fbioe.2023.1189225") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/intestinal-stem-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2014.02.057") Annotation( "DOI:10.1053/j.gastro.2018.08.016") Annotation( "DOI:10.1172/jci.insight.150894") Annotation( "DOI:10.3389/fbioe.2023.1189225") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/intestinal-stem-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Transit amplifying cells (TACs) are an intermediate, undifferentiated population between stem cells and differentiated cells. They can be found in multiple regions such as the small intestine and the colon. TACs of the colon, are integral components of the colonic crypts and vital players in the maintenance of colonic tissue. These cells serve a critical function in the rapid and constant renewal of the epithelium lining the colon, with the whole epithelial surface renewed approximately every 5-7 days. They divide rapidly and progressively differentiate into mature columnar epithelium cells, including enterocytes, goblet cells, and enteroendocrine cells. The continued proliferation of transit amplifying cells is fundamental to maintain the balance in cell population while preventing tissue degeneration and maintaining a state of homeostasis. However, uncontrolled proliferation and compromised differentiation capacity can contribute to the development of colon cancers.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009011") -AnnotationAssertion(Annotation( "DOI:10.1007/s11894-010-0130-3") Annotation( "DOI:10.1016/j.cell.2014.02.057") Annotation( "DOI:10.1101/gad.1674008") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s11894-010-0130-3") Annotation( "DOI:10.1016/j.cell.2014.02.057") Annotation( "DOI:10.1101/gad.1674008") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Transit amplifying cells (TACs) represent an intermediate population between stem cells and fully differentiated cells, and can be found in multiple regions such as the colon and the small intestine. The small intestine's efficiency in absorbing nutrients, its protective barrier function, and its innate cellular renewal every few days is largely dependent on the role played by these TACs. TACs of the small intestine are primarily present in the crypt-villus structure of the intestine, more specifically in the crypt region. They originate from Lgr5+ intestinal stem cells that reside at the base of the crypts. Following their derivation from stem cells, TACs undergo up to six rounds of rapid division over a 48-72 hour period, effectively amplifying the cell population, hence their name. During this process, they gradually migrate upward along the walls of the crypt from where they differentiate into diverse mature cell types such as enterocytes, goblet cells, and Paneth cells. TACs serve as an important element in the homeostasis and regeneration of the intestinal epithelium, amplifying the pool of cells available for differentiation. They also minimize genetic errors during DNA replication by serving as a 'buffer zone' between the long-lived stem cells and the terminal differentiated cells of the gut lining, thus reducing the potential for propagation of mutation-causing defects. Lastly, TACs play an essential part in gastrointestinal tissue repair following injury or inflammation. Their rapid proliferation and subsequent differentiation abilities often help expedite the wound healing process in the intestinal epithelium.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009012") -AnnotationAssertion(Annotation( "DOI:10.1016/j.celrep.2020.107952") Annotation( "DOI:10.1038/s41575-018-0081-y") Annotation( "DOI:10.1073/pnas.1607327113") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.celrep.2020.107952") Annotation( "DOI:10.1038/s41575-018-0081-y") Annotation( "DOI:10.1073/pnas.1607327113") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Intestinal crypt stem cells of the small intestine are a type of adult stem cell intimately involved in the continuous replenishment of the intestinal epithelium, the innermost layer of the intestine responsible for nutrient absorption. These cells, located within the crypts of Lieberkühn, are the origin of various cell lineages that make up the functional units of the small intestine. They possess self-renewal ability, an essential feature of stem cells, which allows them to maintain a steady population in the small intestine. One of the critical roles of intestinal crypt stem cells is to drive the continual renewal process taking place in the small intestine every 3-5 days. By proliferating intensively, these cells produce transient amplifying (TA) cells that are characterized by quick division and progressive differentiation. These cells eventually differentiate into specialized cell types, encompassing absorptive enterocytes, mucin-secreting goblet cells, hormone-secreting enteroendocrine cells, and Paneth cells, all of which have essential roles in digestion and nutrient absorption in the small intestine. Intestinal crypt stem cells of the small intestine are also play a significant part in injury recovery. Under regular conditions, these cells primarily exist in an active state, facilitating the constant renovation of the gut lining. However, upon injury or loss of regular intestinal crypt stem cells reserve intestinal stem cells, a slow-cycling and radio-resistant population, can be stimulated to take over the duties of active crypt stem cells. Such plasticity provides a powerful regenerative mechanism that ensures the intestinal epithelium's function and structural integrity amidst diverse conditions.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009017") -AnnotationAssertion(Annotation( "DOI:10.1016/j.immuni.2022.08.005") Annotation( "DOI:10.1016/j.jcmgh.2021.08.021") Annotation( "DOI:10.1038/s41575-019-0172-4") Annotation( "DOI:10.1038/s41575-023-00769-0") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.immuni.2022.08.005") Annotation( "DOI:10.1016/j.jcmgh.2021.08.021") Annotation( "DOI:10.1038/s41575-019-0172-4") Annotation( "DOI:10.1038/s41575-023-00769-0") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Colon macrophages are a heterogeneous population of macrophages in the gastrointestinal tract. Intestinal macrophages represent the largest group of macrophages in the body and play a key role as sentinels for pathogen recognition and elimination. Because the gastrointestinal tract is continually exposed to a high antigenic load derived from microbes and food intake, macrophages in the intestines are crucial not only for the gut’s immune defense but also for maintaining gastrointestinal homeostasis, avoiding chronic inflammation despite constantly facing foreign antigens. The phenotypic profiles and cytokine production of intestinal macrophages therefore differ from conventional macrophages elsewhere in the body. Colon macrophages orchestrate a wide variety of immune responses. They interact with the colon’s microflora thanks to their location in the mucosal layer, thus playing an important role in shaping gut immunity. By presenting antigens, they help to activate and steer an appropriate immune response either by triggering inflammation against harmful pathogens or by supporting tolerance for beneficial microbes. Therefore, disturbances in the colon macrophage population is thought to contribute to colon-related diseases such as inflammatory bowel disease and colon cancer. These immune cells are versatile and multifunctional, not just limited to defensive actions. Colon macrophages interact with the enteric nervous system to regulate gut secretion and motility. They also play a crucial role in maintaining colon tissue health and integrity by contributing to tissue repair processes. This occurs either through phagocytosis of dead cells and remnants, or indirectly through the release of growth factor molecules, which stimulate cellular proliferation and differentiation necessary for tissue regeneration.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009038") -AnnotationAssertion(Annotation( "DOI:10.1038/mi.2015.32") Annotation( "DOI:10.1038/mi.2016.132") Annotation( "DOI:10.1038/nrgastro.2013.35") Annotation( "DOI:10.1038/s41385-018-0039-y") Annotation( "DOI:10.1073/pnas.1006451107") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/mi.2015.32") Annotation( "DOI:10.1038/mi.2016.132") Annotation( "DOI:10.1038/nrgastro.2013.35") Annotation( "DOI:10.1038/s41385-018-0039-y") Annotation( "DOI:10.1073/pnas.1006451107") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Colon goblet cells are a subset of intestinal goblet cells that are localized in the epithelial lining of the colon. They are specialized secretory epithelial cells that are recognized by their characteristic ‘goblet’ or flask-like shape and typically have a distinctive appearance due to the accumulation of secretory vesicles in their cytoplasm. The primary function of goblet cells is the secretion of mucus, which is an essential substance for the protection and successful functioning of the intestinal tract. In the colon, goblet cells continuously renew the inner mucus layer, which is attached and impervious to bacteria (the outer layer of the colon is unattached and is the habitat of commensal bacteria) The mucus secreted by goblet cells is rich in glycoproteins known as mucins, especially MUC2, forming a complex web of molecules that comprises the foundational structure of the mucus layer. It traps and eliminates harmful bacteria, while selectively allowing beneficial microbiota to access the epithelial surface and aid in digestion and nutrient absorption. The mucus layer not only provides a provides a protective barrier against pathogen invasion, as well as mechanical damage, and the erosive effects of digestive enzymes, it also acts as a lubricant to facilitate the passage of food material. Colon goblet cells are also involved in regulating the local immune response within the gut, maintaining the delicate balance between necessary immune reactions to harmful pathogens and tolerance to beneficial microbiota and dietary substances. Goblet cells can modulate immune responses by presenting antigens to dendritic cells, as well as through the production of immunomodulatory molecules that can alter the behavior of immune cells. When the inner mucus layer of the colon is defective, it might be a pathophysiological mechanism for colitis and infectious diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009039") -AnnotationAssertion(Annotation( "DOI:10.1152/ajpgi.00073.2017") Annotation( "DOI:10.1371/journal.ppat.1010318") Annotation( "DOI:10.3389/fimmu.2022.822867") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/ajpgi.00073.2017") Annotation( "DOI:10.1371/journal.ppat.1010318") Annotation( "DOI:10.3389/fimmu.2022.822867") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Tuft cells, also often referred to as brush cells or caveolated cells, are highly specialized sensory cells found in the colon, among other organs. They were named for their unique appearance under an electron microscope, which presents a distinctive ‘tuft-like’ morphology. They are characterized by apical microvilli, arranged in an irregular tuft that confers a dome-like shape. These cells comprise only a small fraction of the cells in the epithelial layer of the colon, making them among the least populous cell types in this region. Tuft cells in the colon primarily function as chemosensory cells that can sense and respond to environmental changes. These cells can detect and respond to microbial metabolites, helping initiate immune responses against potential threats. Recent studies have revealed that tuft cells play a role in responding to intestinal parasitic infections. When a gastrointestinal parasite infects the colon, tuft cells are activated and release the cytokine IL-25. This action increases the production of tuft cells and triggers essential Th2 immune responses to expel the parasites. Also, the removal of tuft cells has been linked to increased susceptibility to these infections. However, overactivity of tuft cells has been associated with inflammatory bowel disease, highlighting the need for a delicate balance of tuft cell function to maintain colon homeostasis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009041") -AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2013.07.004") Annotation( "DOI:10.1038/s41575-018-0081-y") Annotation( "DOI:10.1073/pnas.1607327113") Annotation( "DOI:10.1111/j.1365-2184.2009.00642.x") Annotation( "DOI:10.1186/s12943-019-0962-x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.cell.2013.07.004") Annotation( "DOI:10.1038/s41575-018-0081-y") Annotation( "DOI:10.1073/pnas.1607327113") Annotation( "DOI:10.1111/j.1365-2184.2009.00642.x") Annotation( "DOI:10.1186/s12943-019-0962-x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Intestinal crypt stem cells of the colon, also known as colon crypt base columnar (CBC) cells, are highly specialized cells primarily responsible for the constant self-renewal of the colonic epithelium. These cells are found in the crypts of Lieberkühn - deeply invaginated sections of the colon's mucosal layer. The prime function of intestinal crypt stem cells of the colon is to serve as the source of constant cell regeneration in the colon. Every few days, these stem cells divide and differentiate into the various other types of intestinal cells, such as enterocytes, goblet cells, and enteroendocrine cells. A constant renewal cycle is necessary due to the harsh environment of the colon where cells continuously encounter abrasive food matter and potential pathogens, leading to a high turnover rate. When the colon's mucosal layer suffers damage, a rapid response is triggered whereby colon crypt stem cells divide faster and are directed to injured sites to repair the epithelial layer. Dysregulation of these cells' function or proliferation can contribute to disorders such as colorectal cancer.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009043") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c190015") Annotation( "DOI:10.1111/aji.12128") Annotation( "DOI:10.1172/jci.insight.163422") Annotation( "DOI:10.1530/REP-09-0147") Annotation( "DOI:10.2174/1570161111311050010") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c190015") Annotation( "DOI:10.1111/aji.12128") Annotation( "DOI:10.1172/jci.insight.163422") Annotation( "DOI:10.1530/REP-09-0147") Annotation( "DOI:10.2174/1570161111311050010") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The endothelial cell of the uterus is a specialized type of endothelial cell found in the inner lining of blood vessels, specifically those in the uterus. These cells play critical roles in maintaining uterine health, fertilization, and pregnancy processes. They form the interior surface of blood vessels, creating a barrier between the vessel lumen and surrounding tissue. This fluid environment, maintained by these endothelial cells, provides a platform for the material exchange between blood and tissues, thereby modulating blood coagulation, immune responses, and controlling vasodilation and vasoconstriction. Functionally, the endothelial cells of the uterus are paramount in regulating the uterine blood flow, an important determinant of successful conception and pregnancy outcomes. They support the vascular changes during the reproductive cycle, particularly the spiral arteriolar development and function, essential in endometrial thickening, maturation, and eventual shedding during menstruation periods. The cells also contribute to angiogenesis, a process crucial during implantation and placenta development, whereby new blood vessels form from pre-existing vessels to supply the growing fetus with oxygen and nutrients. Uterine endothelial cells are also implicated in mediating immune responses within the uterus. Changes in these cells can influence various conditions such as reproductive disorders and complications including heavy menstrual bleeding, endometrial hyperplasia, fertility problems, and cancer.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0009095") -AnnotationAssertion(Annotation( "DOI:10.1007/s00441-021-03471-2") Annotation( "DOI:10.1530/VB-20-0006") Annotation( "DOI:10.3389/fcvm.2018.00101") Annotation( "DOI:10.3389/fphys.2018.00382") Annotation( "DOI:10.3390/ijms20184411") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00441-021-03471-2") Annotation( "DOI:10.1530/VB-20-0006") Annotation( "DOI:10.3389/fcvm.2018.00101") Annotation( "DOI:10.3389/fphys.2018.00382") Annotation( "DOI:10.3390/ijms20184411") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Cardiac endothelial cells are a specialized subset of endothelial cells, the cell type responsible for forming the inner lining of cardiovascular structures such as the heart, and blood vessels. They are a pivotal component of the heart's microenvironment and play a key role in regulating blood pressure, maintaining cardiac homeostasis, in addition to cooperating with other cardiac cells like cardiomyocytes and fibroblasts in the orchestration of a coordinated heart function. Cardiac endothelial cells contribute to the each stage of the heart's operation, whether during relaxation or contraction. Cardiac endothelial cells release nitric oxide, a potent vasodilator that regulates blood vessel dilation consequently controlling blood pressure and flow. They also produce a myriad of growth factors that aid in new blood vessel formation, known as angiogenesis, crucially needed for tissue repair and regeneration when the heart undergoes damage, as in cases of myocardial infarction. Furthermore, cardiac endothelial cells control the passage of nutrients, hormones, and gases between the bloodstream and the heart tissue, ensuring its complex metabolic demands are met adequately. They serve as a selective barrier, regulating the transit of cells and signaling molecules, thereby playing a pivotal role in inflammatory responses and immune cell trafficking. They also contribute to the maintenance of blood fluidity and clotting balance through a complex interplay of anti-thrombic and pro-thrombic factors.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0010008") -AnnotationAssertion(Annotation( "DOI:10.1016/j.exer.2016.03.016") Annotation( "DOI:10.1016/j.pbiomolbio.2003.11.012") Annotation( "DOI:10.1098/rstb.2010.0300") Annotation( "DOI:10.1242/dev.107953") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/lens-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.exer.2016.03.016") Annotation( "DOI:10.1016/j.pbiomolbio.2003.11.012") Annotation( "DOI:10.1098/rstb.2010.0300") Annotation( "DOI:10.1242/dev.107953") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/lens-fiber") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Lens fiber cells are specialized, elongated cells located in the ocular lens, an important part of the eye responsible for focusing light onto the retina to create clear defined images. These cells are unique among body cells, as they are transparent and filled with a clear protein called crystallin to facilitate the passage of light. The entire life cycle of a lens fiber cell includes differentiation from lens epithelial cells, which are the progenitor cells lying at the anterior surface of the lens. In response to various signals, these cells elongate and migrate towards the posterior pole, while simultaneously undergoing a process of denucleation and degradation of cellular organelles. This unique process ensures that the cells do not scatter light, which is critical for the transparency of the lens. After reaching full maturity, lens fiber cells form tightly packed layers, known as laminae, to make up the distinctive architecture of the ocular lens. Lens fiber cells play pivotal roles in vision. Their primary function is to transmit and focus the light that enters the eye onto the retina, providing the sharpness and clarity necessary for vision. The precise alignment and organization of these cells allow for optimal light transmittance and minimizes scattering, thereby maintaining the transparency of the lens. As such, disruptions or abnormalities in lens fiber cells can lead to serious vision impairment, including conditions like cataracts, a common condition characterized by the opacification of the lens.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0011004") -AnnotationAssertion(Annotation( "DOI:10.1002/dvg.23276") Annotation( "DOI:10.1242/dev.193193") Annotation( "DOI:10.3389/fcell.2020.00635/full") Annotation( "https://doi.org/10.1016/j.ydbio.2011.12.042") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/dvg.23276") Annotation( "DOI:10.1242/dev.193193") Annotation( "DOI:10.3389/fcell.2020.00635/full") Annotation( "https://doi.org/10.1016/j.ydbio.2011.12.042") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Neural crest cells are a group of transient and highly migratory cells that originate from the neuroectoderm during the early stages of embryonic development. They are multipotent cells with an exceptional degree of plasticity, capable of differentiating into various somatic cell types and therefore play a fundamental role in the formation of various organs and tissues, making them critical contributors to the developing embryo. After the initiation of neurulation (the formation of the neural tube) neural crest cells start to undergo epithelial-to-mesenchymal transition and delaminate and migrate from the dorsal neural tube to several regions throughout the embryo. They differentiate into a range of diverse cell types, such as neurons and glial cells of the peripheral nervous system, including sensory and autonomic neurons. They also contribute to the formation of adrenal glands, pigment cells in the skin (melanocytes), cardiac structures, including parts of the heart septum and major arteries, as well as bones and cartilage of the face and skull. Disorders or aberrations in the development or migration of the neural crest cells can lead to serious congenital malformations, such as neurocristopathies, including Hirschsprung disease, neuroblastoma, and neurofibromatosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0011012") -AnnotationAssertion(Annotation( "DOI:10.1016/j.diff.2016.05.007") Annotation( "DOI:10.1186/s13578-021-00579-4") Annotation( "DOI:10.1186/s13578-022-00856-w") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.diff.2016.05.007") Annotation( "DOI:10.1186/s13578-021-00579-4") Annotation( "DOI:10.1186/s13578-022-00856-w") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Skeletal muscle fibroblasts (SMFs), with their spindle-shaped morphology, play a crucial role in maintaining the structure and function of skeletal muscles. They produce connective tissues enveloping muscle fibers, offering vital structural support for optimal muscle contraction. SMFs are integral components of the dynamic and complex microenvironment of muscle tissue, contributing significantly to healthy muscle development and homeostasis. SMFs primarily synthesize and secrete extracellular matrix (ECM) components, such as collagen, fibronectin, and proteoglycans. The resulting ECM exhibits mechanical properties that resist tensile forces from muscle contractions, ensuring muscle efficiency. It also crucially transmits force during muscle movement, aids muscle attachment to the skeletal structure, and serves as a scaffold for muscle regeneration and repair. Following injury, they proliferate and transform into myofibroblasts, aiding wound contraction and depositing new ECM for tissue repair. Additionally, these fibroblasts modulate inflammatory responses by secreting cytokines and interact with other cell types, like immune cells and satellite cells, to coordinate muscle healing. In diseases, an imbalance in SMF activity may lead to conditions such as fibrosis, marked by excessive ECM deposition and impaired muscle function.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0011027") -AnnotationAssertion(Annotation( "DOI:10.1007/s00018-019-03104-6") Annotation( "DOI:10.1093/biolre/ioy070") Annotation( "DOI:10.1155/2014/579279") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK53245/") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/placenta-development") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00018-019-03104-6") Annotation( "DOI:10.1093/biolre/ioy070") Annotation( "DOI:10.1155/2014/579279") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK53245/") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/placenta-development") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The chorionic trophoblast cell, vital for placental development in mammals, originates from the blastocyst's outer layer, the trophectoderm. As a significant component of the placenta, the chorion forms the external fetal-maternal interface and consists primarily of chorionic trophoblast cells. These cells are pivotal in embryogenesis, fetal nutrition, waste removal, and immune response generation, contributing crucially to the success of pregnancy. Chorionic trophoblast cells undergo trophoblast differentiation, giving rise to specialized sub-types: cytotrophoblasts and syncytiotrophoblasts. Cytotrophoblasts, found in the inner layer, serve as stem cells and contribute to proliferative activities. In contrast, syncytiotrophoblasts, originating from cytotrophoblasts, form the outer layer with a distinctive multinucleated, continuous, and non-proliferative structure. These cells play a key role in maternal-fetal gas and nutrient exchange, producing essential hormones like human chorionic gonadotropin (hCG), progesterone, and estrogen, vital for sustaining pregnancy. Apart from their physiological roles, chorionic trophoblast cells play a crucial role in shielding the fetus from the maternal immune system. They accomplish this by modifying the expression of major histocompatibility complex (MHC) molecules, which are self-identifying proteins utilized by the immune system to identify foreign cells. Through selective expression of specific types of these molecules, trophoblast cells can evade cytotoxic T cells that might otherwise attack them, while still retaining the ability to attract natural killer cells. These natural killer cells assist in trophoblast invasion and the remodeling of blood vessels in the uterus.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0011101") -AnnotationAssertion(Annotation( "DOI:10.1038/s41586-018-0393-7") Annotation( "DOI:10.1146/annurev-pathol-042420-094031") Annotation( "DOI:10.1172/JCI171268") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41586-018-0393-7") Annotation( "DOI:10.1146/annurev-pathol-042420-094031") Annotation( "DOI:10.1172/JCI171268") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The pulmonary ionocyte is a relatively newly identified, specific epithelial cell type found primarily in the pulmonary or respiratory system. Discovered through novel mapping techniques in 2018, these cells are surprisingly rare, making up less than 2% of the cells in the lung's airway, yet they play an essential role in the airway surface liquid and mucus regulation, a crucial factor in lung health. The main responsibility of the pulmonary ionocyte pertains to the regulation and mobilization of chloride ions. They express a high level of CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) gene, which encodes a protein channel across the membrane of cells that produce mucus, sweat, saliva, tears, and digestive enzymes. The activity of CFTR regulates the movement of chloride ions and fluids in and out of cells, which helps maintain a balance of fluid in the organs they are expressed in. If CFTR is dysfunctional, it can cause conditions such as cystic fibrosis, characterized by thick, sticky mucus that can clog the lungs and obstruct the pancreas, leading to respiratory and digestive issues. The discovery of this cell type offers new doors to the treatment and further understanding of diseases like cystic fibrosis. Increased understanding of pulmonary ionocytes could contribute to the development of novel therapeutic approaches to manipulate the function of CFTR in the lungs and other organs affected by dysfunctional CFTR.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0017000") -AnnotationAssertion(Annotation( "DOI:10.1016/j.resp.2007.06.017") Annotation( "DOI:10.1113/jphysiol.2003.052779") Annotation( "DOI:10.1146/annurev.ph.52.030190.000525") Annotation( "DOI:10.1152/ajplung.00068.2019") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.resp.2007.06.017") Annotation( "DOI:10.1113/jphysiol.2003.052779") Annotation( "DOI:10.1146/annurev.ph.52.030190.000525") Annotation( "DOI:10.1152/ajplung.00068.2019") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Serous cells of the tracheobronchial are found on the surface epithelium, submucosal glands, or both, depending on species. They appear in tubulo-acinar arrangements in submucosal glands. Tracheobronchial serous cells play a vital role in maintaining respiratory health by secreting serous fluid, which is rich in enzymes, ions, and antimicrobial proteins. This serous fluid is essential in clearing out debris, trapping and neutralizing inhaled pathogens and particulate matters, thereby helping in preventing infections. Their secretory products also include water and electrolytes that, combined with mucus (from mucous cells), maintain proper humidity within the airway and lubricate its surfaces, facilitating unhindered airflow. Dysfunction of these cells may lead to diseases such as cystic fibrosis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0019001") -AnnotationAssertion(Annotation( "DOI:10.1007/978-3-540-79090-7_5") Annotation( "DOI:10.1152/japplphysiol.00950.2012") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556044/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/978-3-540-79090-7_5") Annotation( "DOI:10.1152/japplphysiol.00950.2012") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556044/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Tracheobronchial smooth muscle cells are universally located in the tracheobronchial tree and play a critical role in controlling and modulating the size of the airway lumen, thereby contributing significantly to the airflow resistance. These cells display both phasic and tonic characteristics, which are vital for airway contractility. Phasic activity manifests as cyclic contraction and relaxation, similar to the behavior seen in gastrointestinal smooth muscle cells, whilst tonic activity resembles vascular smooth muscle cells, maintaining a constant degree of tension. These features enable tracheobronchial smooth muscle cells to constrict the airways in response to various mechanical and chemical stimuli, including allergens, parasympathetic nervous stimulation, changes in gas composition, and cold air, thus protecting the delicate alveoli from potential damage. Tracheobronchial smooth muscle cells, beyond their contractile function, actively contribute to airway remodeling in conditions like asthma. Their proliferative and synthetic capabilities lead to smooth muscle hypertrophy, hyperplasia, and the production of extracellular matrix components, collectively causing abnormal airway narrowing.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_0019019") -AnnotationAssertion(Annotation( "DOI:10.1038/s41385-018-0039-y") Annotation( "DOI:10.1038/s41385-020-00370-7") Annotation( "DOI:10.1159/000512268") Annotation( "DOI:10.1513/AnnalsATS.201802-128AW") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553208/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41385-018-0039-y") Annotation( "DOI:10.1038/s41385-020-00370-7") Annotation( "DOI:10.1159/000512268") Annotation( "DOI:10.1513/AnnalsATS.201802-128AW") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK553208/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Lung goblet cells are critical components of the respiratory tract, specifically found in the bronchial segments. They are secretory epithelial cells known for their signature \"goblet\" or cup-like shape. Their primary function is to produce and secrete mucus that aids in trapping airborne particles and pathogens, preventing them from reaching the delicate environment of the lung. As part of the lung's epithelial lining, they act as frontline defenders, maintaining lung health and function. The lung goblet cells are densely packed with granules containing mucin glycoproteins, the primary component of mucus. As mucus is produced and secreted, it moves towards the lumen of the lungs where the cilia, hair-like structures of the neighboring ciliated epithelial cells, help to navigate it upwards and out of the respiratory tract. This coordinated action ensures the expulsion of unwanted particles and pathogens, effectively cleaning the respiratory tract. Dysfunction or abnormal proliferation of lung goblet cells can result in pathological conditions such as chronic obstructive pulmonary disorder (COPD) and asthma, where excessive mucus production leads to airway obstruction. Furthermore, lung goblet cells respond to a variety of stimuli, including toxins, allergens, irritants, and infections, adjusting their mucus production accordingly.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000143") -AnnotationAssertion(Annotation( "DOI:10.1016/bs.ctdb.2018.12.002") Annotation( "DOI:10.1016/j.devcel.2020.09.024") Annotation( "DOI:10.1242/dmm.046920") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/bs.ctdb.2018.12.002") Annotation( "DOI:10.1016/j.devcel.2020.09.024") Annotation( "DOI:10.1242/dmm.046920") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Lung neuroendocrine cells, also commonly known as pulmonary neuroendocrine cells (PNECs), are predominantly located in the respiratory epithelium of the bronchial and bronchiolar airways in the lungs. These cells, characterised by their small size and granular appearance, have a distinctive morphology that sets them apart from other lung cells. They are considered part of the diffuse neuroendocrine system due to their scattered distribution through the epithelium and have been classified into solitary cells and clustered forms known as neuroepithelial bodies. The primary function of PNECs is linked to regulation and maintenance of the lung environment. They are sensory in nature and can secrete various bioactive substances such as serotonin, calcitonin, calcitonin gene-related peptides, and bombesin-like peptides which modulate airway smooth muscle tone and influence gut motility. For example, they act as oxygen sensors in response to hypoxia and are responsible for releasing neuropeptides that can induce responses. Moreover, PNECs provide an afferent function as they are equipped with long microvilli that project into the lumen of the bronchus and react to changes in the chemical composition of the luminal content.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000223") -AnnotationAssertion(Annotation( "DOI:10.1007/978-981-13-5895-1_1") Annotation( "DOI:10.1038/nrgastro.2012.168") Annotation( "DOI:10.1038/s41575-020-0271-2") Annotation( "DOI:10.1053/j.gastro.2016.02.030") Annotation( "DOI:10.1111/j.1582-4934.2008.00352.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/978-981-13-5895-1_1") Annotation( "DOI:10.1038/nrgastro.2012.168") Annotation( "DOI:10.1038/s41575-020-0271-2") Annotation( "DOI:10.1053/j.gastro.2016.02.030") Annotation( "DOI:10.1111/j.1582-4934.2008.00352.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Smooth muscle cells of the small intestine form a key component of the enteric system which is imperative for gastrointestinal motility. These cells are a specialized subgroup of smooth muscle cells, specifically found in the layers of the small intestine. The small intestine represents a significant part of the digestive system and plays a crucial role in the absorption of nutrients into the bloodstream. Smooth muscle cells of the small intestine are predominantly responsible for creating the contractions and relaxations that constitute intestinal motility. They trigger these movements in a coordinated and rhythmic manner known as peristalsis and segmentation. Peristalsis is a form of longitudinal and sequential contraction that pushes food from the upper gastrointestinal tract to the lower parts. Segmentation works by squeezing the intestine randomly, thereby facilitating the mixing of food particles and increasing contact with absorptive surfaces. These two processes are critical for intestinal motility and an effective digestive process. Apart from facilitating transit of food through contractions, these smooth muscle cells play a role in controlling the diameter of the vessels and the blood flow in the small intestine by their constrictive ability. These smooth muscle cells also contribute to the structural integrity of the small intestine by forming a part of its muscularis externa and muscularis mucosae. Additionally, they interact with other cell types such as enteric neurons and interstitial cells of Cajal (ICC), forming an integrated cellular network that ensures optimal functioning of the intestinal tract.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000275") -AnnotationAssertion(Annotation( "DOI:10.1152/advan.00025.2003") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532857/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556137/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/advan.00025.2003") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK532857/") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK556137/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The smooth muscle fiber of the ileum primarily refers to a specific class of muscle cells present in the ileum, the final segment of the small intestine. Distinguished from skeletal muscle and cardiac muscle, smooth muscle does not possess striations and is under the control of the involuntary nervous system. These cells primarily facilitate the movement of digested food, a process known as peristalsis. These cells coordinate rhythmic contractions and relaxations, propelling the chyme toward the colon. This process is vital for efficient nutrient absorption in the small intestine and the transfer of undigested material to the large intestine. Moreover, the smooth muscle fibers enhance the ileum's ability to expand and adapt to volume changes without elevating internal pressure, known as compliance. The smooth muscle fiber of the ileum contributes to reflex responses, enhancing muscle contractions to propel intestinal contents in different physiological conditions. This involves intricate molecular signaling with neurotransmitters, hormones, and local signals. Thus, beyond its structural role, the smooth muscle fiber is crucial for the physiological functions of digestion and absorption in the small intestine.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000278") -AnnotationAssertion(Annotation( "DOI:10.1016/j.celrep.2018.11.086") Annotation( "DOI:10.1038/s41385-022-00565-0") Annotation( "DOI:10.1089/ten.teb.2016.0352") Annotation( "DOI:10.1101/2020.02.19.937615v2.full") Annotation( "DOI:10.1152/ajprenal.00327.2009") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.celrep.2018.11.086") Annotation( "DOI:10.1038/s41385-022-00565-0") Annotation( "DOI:10.1089/ten.teb.2016.0352") Annotation( "DOI:10.1101/2020.02.19.937615v2.full") Annotation( "DOI:10.1152/ajprenal.00327.2009") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Epithelial cells of the urethra constitute the inner lining of the urethra, forming a mucosal barrier that helps protect the body from external contaminants. The type of epithelial cells found in the urethra can vary along its length. In general, the urethra is lined with stratified squamous epithelium in its distal or external portion, which is closer to the external environment. This type of epithelium provides protection against mechanical stress and pathogens. Closer to the bladder, the urethra transitions to a different type of epithelium. In males, the proximal part of the urethra is lined with pseudostratified columnar epithelium in the region where it passes through the prostate gland. In females, the transitional epithelium may be present in the proximal part of the urethra near the bladder. Epithelial cells of the urethra are primarily dedicated to the protection of underlying tissues from mechanical stress and pathogens during micturition (urination). They do this by creating a tight, impermeable barrier that prevents the entry and colonization of invading microbes. They produce a variety of antimicrobials and are tightly packed to prevent infiltration between cells, forming an integral part of the body's innate immune system. In addition to their protective role, these cells are involved in the secretion of mucus. This mucus acts as a lubricant that ensures the smooth and easy passage of urine from the bladder through the urethra and out of the body, reducing any potential damage from the erosive action of urine. Epithelial cells of the urethra are dynamic, constantly renewing their numbers to replace any cells lost through wear and tear. Any disruptions to the function or integrity of these cells may contribute to urinary tract infections and other urological disorders.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000296") -AnnotationAssertion(Annotation( "DOI:10.1016/S0165-6147(00)01600-X") Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1080/07853890600585795") Annotation( "DOI:10.2147/COPD.S38938") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S0165-6147(00)01600-X") Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1080/07853890600585795") Annotation( "DOI:10.2147/COPD.S38938") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Bronchial goblet cells are a subset of goblet cells that are predominantly found in the respiratory tract, specifically within the bronchi and bronchioles. Named for their flask-like shape and their apparent similarity to a goblet, these cells contribute significantly to the maintenance of lung health and homeostasis. In humans, goblet cells comprise up to 25% of the bronchial epithelial columnar cells and are an integral part of the mucus-secreting system of the respiratory tract and are primarily responsible for the production and secretion of mucus. The mucus secreted by bronchial goblet cells serves as a protective layer over the delicate and sensitive structures of the bronchi. The mucus, an amalgamation of glycoproteins, lipids, and other substances, traps any inhaled dust, microorganisms, and pollutants, preventing them from reaching deeper lung tissues and causing potential damage. Additionally, the mucus secretion aids in moistening the airway surface, thereby facilitating effective gas exchange and maintaining the overall health of the respiratory tract. Bronchial goblet cells have an innate defense mechanism wherein they increase secretion of mucus in response to irritants, infection or inflammation in the airway. This strategy, however, if incessantly stimulated, can lead to airway obstruction and health complications like asthma and chronic obstructive pulmonary disease (COPD).") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000312") -AnnotationAssertion(Annotation( "DOI:10.1038/mi.2015.32") Annotation( "DOI:10.1042/BSR20201471") Annotation( "DOI:10.1111/imr.12182") Annotation( "DOI:10.1146/annurev-physiol-021115-105447") Annotation( "DOI:full/10.1111/febs.15731") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/mi.2015.32") Annotation( "DOI:10.1042/BSR20201471") Annotation( "DOI:10.1111/imr.12182") Annotation( "DOI:10.1146/annurev-physiol-021115-105447") Annotation( "DOI:full/10.1111/febs.15731") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The large intestine goblet cell is a highly specialized type of mucosal epithelial cell that is located within the epithelial lining of the large intestine. The key function of goblet cells is the production and secretion of a protective layer of mucus which is essential for maintaining gut homeostasis. Goblet cells have a characteristic 'goblet-like' shape, which adapts them for this specific function: organelles such as the nucleus, mitochondria, ER, and Golgi apparatus are located in the basal portion of the cell; while the vesicles with mucins (required for mucus production) are located apically, close to the apical membrane where their exocytosis takes place. The goblet cells of the large intestine produce a thick layer of mucus that coats the intestinal lumen. This mucus provides a barrier that prevents the direct contact of intestinal cells with bacteria and other potentially toxic substances present in the digestive tract. It also lubricates the intestine to facilitate the smooth passage of digested food material. It is worth noting that the mucus system differs between the small and large intestine: although in both the mucus is built around MUC2 mucin polymers produced by goblet cells, it is becoming clear that there are several types of goblet cells that function in different ways. From an immunological perspective, large intestine goblet cells participate in promoting a balanced gut immune response. They contribute to the process known as immune tolerance, where they prevent the body's immune system from overreacting to the trillions of bacteria present in the gut. These cells achieve this by secreting molecules that help maintain a calm environment, limiting inflammatory reactions unless absolutely necessary. Any dysfunction or decrease in the number of goblet cells can result in a compromised intestinal barrier leading to various health issues, including inflammatory bowel disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000320") -AnnotationAssertion(Annotation( "DOI:10.1007/s11894-010-0131-2") Annotation( "DOI:10.1038/nature10863") Annotation( "DOI:10.1038/s41575-022-00675-x") Annotation( "DOI:10.1111/imr.12182") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s11894-010-0131-2") Annotation( "DOI:10.1038/nature10863") Annotation( "DOI:10.1038/s41575-022-00675-x") Annotation( "DOI:10.1111/imr.12182") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The ileal goblet cell is a subset of intestinal goblet cells that are a key component of the epithelium of the ileum, a portion of the small intestine. Goblet cells are named and characterized by their distinctive 'goblet' shape with a narrow base tapering to a broad apical surface; located between the lumen of the intestine and the internal environment of the body, they serve an integral role in maintaining gut homeostasis. Like all intestinal goblet cells, the primary function of ileal goblet cells is the production and secretion of mucins, large, heavily glycosylated proteins that combine to form mucus. This mucus lining serves as a protective barrier, shielding the underlying tissue from the potentially damaging effects of digestive enzymes, acids, or pathogenic microbes present in the gut lumen. Their strategic location in the ileal segment of the intestinal tract aids in efficient nutrient absorption while also maintaining an impregnable barrier against potential pathogens, ensuring an intricate balance. Goblet cells of the ileum also play a significant role in immune modulation. They are thought to promote oral tolerance (the immune unresponsiveness to orally ingested antigens such as food) by delivering luminal antigens to underlying dendritic cells, thereby driving immune education and tolerogenic responses. They also secrete bioactive molecules, such as the trefoil factor peptide TFF3, which aids in repairing damaged epithelium. By performing these crucial functions, ileal goblet cells not only aid in maintaining intestinal health, but also contribute significantly to the overall well-being of the organism.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000326") -AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1164/ajrccm.154.6.8970383") Annotation( "DOI:10.1165/ajrcmb.25.5.f218") Annotation( "DOI:10.2147/COPD.S38938") Annotation( "DOI:10.3109/01902148.2013.791733") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(02)00083-3") Annotation( "DOI:10.1164/ajrccm.154.6.8970383") Annotation( "DOI:10.1165/ajrcmb.25.5.f218") Annotation( "DOI:10.2147/COPD.S38938") Annotation( "DOI:10.3109/01902148.2013.791733") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Tracheal goblet cells are specialized cells found in the lining of the trachea. They are a key component of the tracheobronchial epithelium, a critical region responsible for maintaining open airways and preventing the ingress of harmful particles or irritants. Goblet cells are filled with membrane-bound secretory granules, which are responsible for their distinctive, goblet-like shape. Goblet cells specialize in the production and secretion of mucus, a slimy substance composed primarily of glycoproteins (mucins) and water. This sticky mucus serves to trap foreign particles and pollutants, such as dust, bacteria, and viruses that an organism breathes in. After trapping these particles, the cilia on neighboring ciliated epithelial cells beat synchronously, forming a coordinated escalator to push mucus upwards toward the pharynx, where it's either swallowed or expectorated. This system, known as the mucociliary clearance or escalator, forms a crucial barrier in the lungs against respiratory pathogens and external insults. Changes in the number or function of tracheal goblet cells can engender health complications. For example, goblet cell hyperplasia, or excessive growth of goblet cells, is a common feature of chronic inflammatory lung diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). This condition results in mucus hypersecretion, airway blockage, and increased risk of respiratory tract infections.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000329") -AnnotationAssertion(Annotation( "DOI:10.1002/dvdy.24250") Annotation( "DOI:10.1016/B978-012330215-1/50029-6") Annotation( "DOI:10.1016/S0034-5687(01)00214-") Annotation( "DOI:10.1074/jbc.M208826200") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/dvdy.24250") Annotation( "DOI:10.1016/B978-012330215-1/50029-6") Annotation( "DOI:10.1016/S0034-5687(01)00214-") Annotation( "DOI:10.1074/jbc.M208826200") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Serous cells are specialized epithelial cells and can be found lining the trachea and brochus. They are among the major components of the respiratory epithelium. These cells are known for their production and release of serous secretions, which form a key component of airway mucus. These secretions primarily comprise of water, proteins, and enzymes, such as lysozyme and peroxidases. Their main purposes are to provide hydration and lubrication to the airway tissue and to act as a defense mechanism against pathogens. While the lysozyme and peroxidases contribute to the destruction of bacteria, the water in the secretions helps to liquefy the mucus, facilitating the effective functioning of the ciliated cells that propel the mucus up the trachea. The coordinated action between serous cells and other cell types in the tracheal epithelium proves essential for maintaining a healthy respiratory tract. Dysfunction or damage of these cells can lead to various respiratory tract disorders, such as cystic fibrosis and asthma.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000330") -AnnotationAssertion(Annotation( "DOI:10.1152/physrev.00039.2014") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK534789/") Annotation( "https://www.sciencedirect.com/topics/neuroscience/secretory-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/physrev.00039.2014") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK534789/") Annotation( "https://www.sciencedirect.com/topics/neuroscience/secretory-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Serous cells of the epithelium of the bronchus, also termed bronchial serous cells, are predominantly found within the seromucous glands located in the bronchial submucosa. Serous cells can also be found in the trachea and are characterized by small, sparse, electron-dense granules. Serous cells are specialized cells responsible for the production and secretion of serous fluid. Serous fluid, mainly composed of water, proteins, and various types of salts, provides many benefits: it helps moisturize the airways, keep the bronchial tube walls moist, and safeguard the bronchial tubes against various foreign particles and infectious agents such as bacteria and viruses. Moreover, the fluid released by these cells assists in the lubrication and reduced friction in the bronchi, allowing for smoother airflow.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000331") -AnnotationAssertion(Annotation( "DOI:10.1038/nrgastro.2013.35") Annotation( "DOI:10.1038/nri3738") Annotation( "DOI:10.1084/jem.20191130") Annotation( "DOI:10.3389/fphys.2021.699152/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nrgastro.2013.35") Annotation( "DOI:10.1038/nri3738") Annotation( "DOI:10.1084/jem.20191130") Annotation( "DOI:10.3389/fphys.2021.699152/full") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Enterocytes of the epithelium of the small intestine are specialized cells that reside in the lining of the small intestine, and are primarily responsible for the essential process of nutrient absorption. These cells are columnar epithelial cells with an apical surface lined with microvilli, a feature referred to as the 'brush border', to maximize the surface area available for absorption. Enterocytes play a critical role in both the digestion and absorption of nutrients from food. Their extensive brush border contains enzymes that further assist in nutrient breakdown and transport proteins that transfer nutrients, such as glucose, amino acids, lipids, and vitamins, across the cell membrane. The enterocytes of the small intestine also participate in the barrier function of the gut lining. Enterocytes are connected by tight junctions, which act as a primary defense line against pathogenic invasion by maintaining intestinal barrier integrity. Additionally, their cell surface is coated in glycocalyx and mucus which forms a defensive barrier preventing the penetration of harmful bacteria into the systemic circulation.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000334") -AnnotationAssertion(Annotation( "DOI:10.1038/nri3738") Annotation( "DOI:10.1084/jem.20191130") Annotation( "DOI:10.1111/j.1365-2249.2011.04523.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nri3738") Annotation( "DOI:10.1084/jem.20191130") Annotation( "DOI:10.1111/j.1365-2249.2011.04523.x") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Enterocytes of the epithelium proper of the ileum, commonly known as ileal enterocytes, are specialized epithelial cells found lining the inner surface of the ileum, the final section of the small intestine in the human body. They play a pivotal role in nutrient absorption, digestive metabolic functions, and the maintenance of the host’s immune response. Like enterocytes in other parts of the intestine, ileal enterocytes exhibit distinct characteristics specific to their function and role. They have microvilli on their apical surfaces to increase absorption and are important in the absorption of vitamins and the reabsorption of bile salts. These cells also produce enzymes that metabolize lipids and xenobiotics.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000342") -AnnotationAssertion(Annotation( "DOI:10.1007/s00018-002-8412-z") Annotation( "DOI:10.1038/nature09637") Annotation( "DOI:10.1038/nrmicro2546") Annotation( "DOI:10.1146/annurev-physiol-030212-183744") Annotation( "DOI:10.3389/fimmu.2020.00587") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00018-002-8412-z") Annotation( "DOI:10.1038/nature09637") Annotation( "DOI:10.1038/nrmicro2546") Annotation( "DOI:10.1146/annurev-physiol-030212-183744") Annotation( "DOI:10.3389/fimmu.2020.00587") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Paneth cells of the epithelium of the small intestine, often referred to simply as Paneth cells, are a type of specialized secretory cell that forms an integral part of the intestinal crypts of Lieberkühn – gland-like invaginations lining the small intestine. Named after the Austrian physician Josef Paneth, who first identified them in the late 19th century, these cells are significant because of their substantial involvement in the mucosal immune system and intestinal stem cell maintenance. Functionally, Paneth cells play a key role in maintaining gut homeostasis and in the first line of antimicrobial defense, mainly through the secretion of a range of antimicrobial peptides and proteins. These include lysozymes, cryptdins, or alpha-defensins, which have potent activity against various gut pathogens including bacteria, fungi, and parasites. Furthermore, Paneth cells of the small intestine are also known to release secretory phospholipase A2, an enzyme active against gram-positive bacteria. Additionally, they secrete several growth factors like EGF, TGF-alpha, and Wnt3, which are crucial for stem cell support and the overall maintenance of intestinal mucosal integrity. The unique location of Paneth cells at the base of the crypts positions them in very close proximity to intestinal stem cells. This not only facilitates their role in stem cell maintenance but also involves them in epithelial regeneration and repair following injury. Dysfunctional Paneth cells have been associated with several gut disorders, like Crohn’s disease and necrotizing enterocolitis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000343") -AnnotationAssertion(Annotation( "DOI:10.1007/s11894-010-0130-3") Annotation( "DOI:10.1038/nrgastro.2013.35") Annotation( "DOI:10.1038/nri3738") Annotation( "DOI:10.1084/jem.20191130") Annotation( "DOI:10.3389/fimmu.2019.00277") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s11894-010-0130-3") Annotation( "DOI:10.1038/nrgastro.2013.35") Annotation( "DOI:10.1038/nri3738") Annotation( "DOI:10.1084/jem.20191130") Annotation( "DOI:10.3389/fimmu.2019.00277") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Enterocytes of the colon are specialized epithelial cells located in the lining of the colon, the largest part of the large intestine. These cells play a critical role in absorbing water, electrolytes, and certain vitamins from the food material passed on from the small intestine. With a unique structure of finger-like protrusions referred to as microvilli, the enterocytes increase their surface area for effective absorption. The colon is the last part of the digestive system, and as such, it is responsible for compacting undigested food materials and forming fecal matter. Enterocytes of the colon facilitate this process effectively through absorption of water. Enterocytes are known for their high regeneration potential, replenishing every 4-5 days, enabling the healthy functioning of the colon. They originate from stem cells located in the crypt of the colon and differentiate into mature enterocytes as they migrate upwards towards the luminal surface. This constant turnover aids in maintaining the intestinal barrier, preventing the entry of detrimental substances into the systemic circulation. Their tight junctions with other epithelial cells provide a robust barrier against invasive pathogens. Enterocytes of the colon are involved in the communication with the gut microbiota. These cells harbor enzymes necessary for the metabolism of short-chain fatty acids, which are the byproducts of the fermentation process by gut bacteria. Short-chain fatty acids serve as a major energy source for colonocytes and are important for maintaining colonic health. The dysfunction of enterocytes, therefore, could lead to disorders such as inflammatory bowel disease or colorectal cancer.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000347") -AnnotationAssertion(Annotation( "DOI:10.1164/rccm.201408-1492PP") Annotation( "DOI:10.1165/rcmb.2021-0150ED") Annotation( "DOI:10.1242/dmm.006031") Annotation( "DOI:10.3389/falgy.2021.787128") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1164/rccm.201408-1492PP") Annotation( "DOI:10.1165/rcmb.2021-0150ED") Annotation( "DOI:10.1242/dmm.006031") Annotation( "DOI:10.3389/falgy.2021.787128") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Basal cells of epithelium of trachea are a vital cell population within the respiratory tract's lining, specifically in the trachea's epithelium. These cells are located at the base of the columnar epithelial cell layer and can be identified by their characteristic cuboidal shape, a large nucleus and few organelles, as well as scattered microvilli. The basal cells are not in direct contact with the lumen, which is covered by a layer of ciliated and non-ciliated columnar cells. Instead, the basal cells are connected to, and line, a thin basement membrane. The primary function of basal cells involves stem cell activity and epithelial tissue maintenance. Basal cells in the trachea play a crucial role in the regeneration and repair of the tracheal epithelium during damage or injury, acting as progenitor cells for ciliated and secretory cells. They serve as a reserve pool of cells, ready to proliferate and differentiate as needed for homeostatic tissue maintenance or in repair processes when the epithelium has been compromised, whether by injury or disease. These cells are also implicated in numerous disease pathways. Conditions like chronic obstructive pulmonary disease and lung cancer exhibit unusual behavior and quantities of tracheal basal cells, making these cells a focal point in respiratory disease research. In summary, basal cells in the tracheal epithelium have a multifaceted role in maintaining, repairing, and influencing tracheal health, playing a crucial role in the stability of the respiratory system.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000348") -AnnotationAssertion(Annotation( "DOI:10.1002/ar.1092380310") Annotation( "DOI:10.1038/labinvest.2015.114") Annotation( "DOI:10.1136/thx.2004.la0104") Annotation( "DOI:10.3389/falgy.2021.787128") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/ar.1092380310") Annotation( "DOI:10.1038/labinvest.2015.114") Annotation( "DOI:10.1136/thx.2004.la0104") Annotation( "DOI:10.3389/falgy.2021.787128") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The basal cells of the epithelium of the bronchus are a vital component of the cellular linings found within the bronchial tubes of the respiratory system. They are typically nonciliated, cuboidal cells that are tightly attached to the basement membrane, thereby providing structural support for the epithelial tissues. The function of the basal cell of the bronchial epithelium is multifaceted, underscoring the importance of these cells for respiratory health. One of their primary roles involves regeneration and cellular turnover. They serve as local stem cells, being able to proliferate and differentiate into other cell types such as ciliated and secretory epithelial cells. This regenerative function of basal cells is required for maintaining the integrity of the bronchial epithelium and is especially critical after injury or during disease states when there is an increased need for new cells to repair damaged tissue or replace lost cells. In addition to these regenerative duties, basal cells of the bronchial epithelium play a crucial role in providing a defensive barrier against inhaled substances. They participate in the coordinated immune response directed against airborne pathogens, foreign particles, and toxins that enter the respiratory tract. Basal cells can respond to inflammation or irritation by altering their activities, which include proliferating, differentiating, or producing bioactive substances. Throughout all these responses, the basal cells help to maintain the homeostasis of the bronchial tubes, allowing for the efficient transport of air to and from the lungs.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000349") -AnnotationAssertion(Annotation( "DOI:10.1038/mi.2013.30") Annotation( "DOI:10.1093/jb/mvv121") Annotation( "DOI:10.1111/j.1574-695X.2007.00359.x") Annotation( "DOI:10.3389/fimmu.2019.01499") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/mi.2013.30") Annotation( "DOI:10.1093/jb/mvv121") Annotation( "DOI:10.1111/j.1574-695X.2007.00359.x") Annotation( "DOI:10.3389/fimmu.2019.01499") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Microfold cells, also widely known as M cells, are a distinct type of epithelial cell found in the gut, including the small intestine, specifically within the Peyer’s patches, a component of the gut-associated lymphoid tissue (GALT). M cells play a crucial role in the immune response within the intestine environment. These cells are characterized by microfolds (short microvilli, which give them their name), a reduced, thin glycocalyx, and a deeply invaginated basolateral membrane next to immune cells such as B cells, T cells, and macrophages, and DCs. @@ -1045,234 +1024,234 @@ These features allow the M cells to form the first point of contact between the Furthermore, M cells have complex interactions with members of the gut microbiota and other immune cells, making them essential for maintaining gut homeostasis. This cross-talk shapes the diversity and makeup of the gut microbiota, subsequently having a substantial impact on the host's health. In certain situations, pathogenic organisms can exploit the transcytosis mechanism to infiltrate the host's system, causing various infectious diseases. The overarching significance of M cells in the small intestine lies in their role of surveillance and protection against a wide range of potentially harmful pathogens, while also facilitating beneficial interactions with commensal microbes.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000353") -AnnotationAssertion(Annotation( "DOI:10.1016/j.jhep.2016.07.009") Annotation( "DOI:10.1038/cmi.2016.5") Annotation( "DOI:10.1038/s41575-018-0020-y") Annotation( "DOI:10.1038/s41575-020-00411-3") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/disse-space") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.jhep.2016.07.009") Annotation( "DOI:10.1038/cmi.2016.5") Annotation( "DOI:10.1038/s41575-018-0020-y") Annotation( "DOI:10.1038/s41575-020-00411-3") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/disse-space") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial cells of hepatic sinusoids, also known as hepatic or liver sinusoidal endothelial cells (HSEC or LSEC), are unique, specialized type of endothelial cells located in the liver. They are the building units of the hepatic sinusoid, which functions as a specialized capillary system that facilitates the exchange of various substances between the blood and hepatocytes. This finely tuned environment, facilitated by HSEC, is primordial for the liver's multiple physiologic functions that include nutrient metabolism, toxin inactivation, and immunomodulation. The HSEC have a distinctive morphology that sets them apart from the typical endothelial cells found in other organs. These cells are exceptionally thin to allow for efficient transfer of molecules and characterized by fenestrations, which are non-diaphragmatic, sieve-like openings that provide an open filtration pathway from the sinusoidal lumen to the space of Disse, where hepatocytes are exposed. The fenestration is a crucial feature that allows lipoproteins, nutrients, and other plasma components easy access to hepatocytes for essential liver functions, including clearance of waste products and metabolic regulation. HSECs further play a vital role in immune functionality in the liver. They act as a sentinel, determining the nature of the immune response to encountered particulates. They possess a capacity for antigen presentation and express a series of immune-related surface molecules, which helps in immunosurveillance and immunoregulation. The HSEC also aid in the removal and destruction of virus-infected cells, harmful microorganisms, and circulatory waste products to safeguard liver health and general systemic cleanliness.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000398") -AnnotationAssertion(Annotation( "DOI:10.1007/s00441-008-0706-5") Annotation( "DOI:10.1007/s10456-021-09785-7") Annotation( "DOI:10.1016/j.ccm.2021.08.005") Annotation( "DOI:10.1016/j.jvs.2004.03.043") Annotation( "DOI:10.1177/153857440303700107") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00441-008-0706-5") Annotation( "DOI:10.1007/s10456-021-09785-7") Annotation( "DOI:10.1016/j.ccm.2021.08.005") Annotation( "DOI:10.1016/j.jvs.2004.03.043") Annotation( "DOI:10.1177/153857440303700107") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Endothelial cells of the artery, also referred to as arterial endothelial cells, form an integral part of the arterial system. They form a single layer, known as the endothelium, lining the interior surface of arteries, and are able to respond to the high-pressure and flow conditions present in arteries. The primary role of these cells is to provide a barrier between the vessel wall and the blood, exhibiting selective permeability to regulate the movement of liquids, gases, and blood-borne substances across the vascular wall. Arterial endothelial cells significantly contribute to maintaining vascular homeostasis. They are at the forefront of sensations and responses to mechanical stimuli, like shear stress and blood pressure changes. An additional key function pertains to the production of nitric oxide, which helps to regulate vascular tone and blood pressure, prevents platelet aggregation, limits leukocyte adhesion to the endothelium, and inhibits smooth muscle cell proliferation. These varied but connected functions help to preclude the development of atherosclerosis, ensuring normal circulation and arterial health. Moreover, these cells play a pivotal role in inflammation and coagulation processes. During inflammatory events, they express various adhesion molecules, aiding in leukocyte recruitment and rolling onto the vessel walls for immune response. They also produce anticoagulant and procoagulant substances, involved in blood clotting and clot dissolution, respectively. Dysregulation of the usual functions of arterial endothelial cells can result in serious pathophysiological conditions, such as atherosclerosis, hypertension, and other cardiovascular diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000413") -AnnotationAssertion(Annotation( "DOI:10.1007/s004290050160") Annotation( "DOI:10.1111/j.1442-9071.2012.02818.x") Annotation( "DOI:10.1167/tvst.8.4.32") Annotation( "DOI:10.2147/OPTH.S26048") Annotation( "DOI:10.3389/fimmu.2022.918619") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s004290050160") Annotation( "DOI:10.1111/j.1442-9071.2012.02818.x") Annotation( "DOI:10.1167/tvst.8.4.32") Annotation( "DOI:10.2147/OPTH.S26048") Annotation( "DOI:10.3389/fimmu.2022.918619") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Epithelial cells of the lacrimal sac play a significant role in the physiology of tear drainage, acting as an integral part of the lacrimal drainage system. The lacrimal sac is part of the nasolacrimal duct system, a conduit which connects the eye to the nasal cavity, and is lined by multilayered, non-keratinizing, squamous epithelial cells. The epithelial cells of the lacrimal sac are specialized for the purpose of maintaining a moist environment and protecting the surface of the eye. They form a barrier that traps and removes potential contaminants from the tear film during the drainage process. These cells also actively contribute to tear turnover by expediting the drainage of excess tears. Dysfunctional epithelial cells of the lacrimal sac can lead to dacryocystitis, a condition characterized by inflammation of the lacrimal sac. Epithelial cells of the lacrimal sac also perform various other tasks which ensure the overall health of the eye. They are involved in the regulation of immune responses within the lacrimal apparatus and may have a possible role in host-microbiome interactions. Thus, epithelial cells of the lacrimal sac play a multifaceted role in tear drainage and ocular surface defence, directly translating to eye health and vision quality.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000436") -AnnotationAssertion(Annotation( "DOI:10.1016/S1350-9462(98)00011-1") Annotation( "DOI:10.1038/srep31171") Annotation( "DOI:10.1111/j.1444-0938.2008.00260.x") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/ciliary-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S1350-9462(98)00011-1") Annotation( "DOI:10.1038/srep31171") Annotation( "DOI:10.1111/j.1444-0938.2008.00260.x") Annotation( "https://www.sciencedirect.com/topics/medicine-and-dentistry/ciliary-muscle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Ciliary muscle cells are specialized contractile cells that constitute the ciliary muscle, a component of the eye in vertebrates. These cells form a ring around the iris, the eye's colored part. Ciliary muscle cells fall into the category of smooth muscle cells, characterized by their lack of the striations typical of skeletal and cardiac muscle cells. Their shape is elongated and pointed at both ends, and they occur in multi-nucleated syncytia where many cells share the same cytoplasm. Ciliary muscle cells primarily control the eye's accommodation, adjusting the thickness and curvature of the lens to fine-tune the focus of light on the retina. When ciliary muscle cells contract, tension on the zonular fibers holding the lens in place is reduced, allowing the lens to become thicker and increase its refractive power. Conversely, when the ciliary muscle cells relax, the tension on the zonular fibers increases, causing the lens to become flatter and decrease its refractive power. This adjustment facilitates clear vision at different distances, ranging from near to far objects. Additionally, ciliary muscle cells play a role in draining the eye's aqueous humor, which is the clear fluid that fills the anterior chamber of the eye between the cornea and the lens. By controlling the shape of the surrounding trabecular meshwork through muscle contractions and relaxations, ciliary muscle cells indirectly regulate intraocular pressure, a factor critical for preventing ocular conditions like glaucoma.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000443") -AnnotationAssertion(Annotation( "DOI:10.1007/978-94-011-2354-9_6") Annotation( "DOI:10.1007/s004670050586") Annotation( "DOI:10.1016/B978-0-323-35775-3.00011-4") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1681/ASN.2012010029") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/978-94-011-2354-9_6") Annotation( "DOI:10.1007/s004670050586") Annotation( "DOI:10.1016/B978-0-323-35775-3.00011-4") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1681/ASN.2012010029") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Epithelial cells of the nephron form the epithelial lining of the nephron, the functional unit of the kidney that filters the blood to produce urine. Each kidney holds approximately one million nephrons, each of which is composed of several distinct parts, including the renal corpuscle, the proximal tubule, the loop of Henle, and the distal tubule. All sections are lined with highly specialized epithelial cells adapted to perform different functions within the sections of the nephron. The different epithelial cells of the nephron play crucial roles in the process of filtration, reabsorption, secretion, and excretion, essential tasks for the regulation of body fluid composition and volume. During filtration, the blood is forced through the walls of glomerular capillaries into the Bowman's capsule, where waste products, electrolytes, and water are separated from the blood cells and proteins. As the filtrate then passes through the nephron, epithelial cells of the tubular portion reabsorb useful substances such as glucose, amino acids, and electrolytes, along with most of the water back into the bloodstream. Waste products like urea and creatinine, as well as excess ions and water, remain in the tubule for elimination. In addition, epithelial cells of the nephron also play a vital part in maintaining the body's acid-base balance by actively secreting hydrogen ions into the urine while reabsorbing bicarbonate from the urine back into the blood. They are also involved in the regulation of blood pressure through the renin-angiotensin-aldosterone system and contribute to erythrocyte production by releasing the hormone erythropoietin in response to low oxygen levels in the blood. Abnormalities or damage in renal epithelial cells often result in impaired kidney function, leading to various renal diseases.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000449") -AnnotationAssertion(Annotation( "DOI:10.1016/j.ajpath.2014.01.014") Annotation( "DOI:10.1016/j.semnephrol.2019.04.005") Annotation( "DOI:10.2215/CJN.05760513") Annotation( "DOI:10.2215/CJN.08880914") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.ajpath.2014.01.014") Annotation( "DOI:10.1016/j.semnephrol.2019.04.005") Annotation( "DOI:10.2215/CJN.05760513") Annotation( "DOI:10.2215/CJN.08880914") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney collecting duct epithelial cells are a specialized type of cells that form an integral part of the renal system. Located in the collecting duct system of the kidneys, these cells are responsible for one of the final steps in the process of urine formation, and they are instrumental in the fine tuning of the volume and composition of urine by reabsorbing water and certain solutes back into the bloodstream. These cells express specific channels and carriers that actively and passively transport ions and water. They also have channels on their membranes, such as sodium channels and potassium channels, involved in reabsorbing or secreting these electrolytes depending upon the body's needs. The function of renal collecting duct epithelial cells can be regulated by a variety of hormones, including vasopressin (antidiuretic hormone), which can modulate the ion channels and carriers and hence indirectly influence body fluid homeostasis. Aside from their function in ion and water balance, kidney collecting duct epithelial cells also aid in maintaining the body's acid-base balance. They have specialized functionality to secret hydrogen ions into the tubular lumen, which contributes to acid excretion. Any dysfunction may have serious implications and lead to various renal or systemic diseases, exemplifying the importance of these cells in maintaining overall body homeostasis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000454") -AnnotationAssertion(Annotation( "DOI:10.1016/j.jinf.2015.06.006") Annotation( "DOI:10.1111/j.1751-1097.2007.00226.x") Annotation( "DOI:10.1615/CritRevEukaryotGeneExpr.2020028454") Annotation( "DOI:10.5114/pdia.2013.33376") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/melanocyte") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.jinf.2015.06.006") Annotation( "DOI:10.1111/j.1751-1097.2007.00226.x") Annotation( "DOI:10.1615/CritRevEukaryotGeneExpr.2020028454") Annotation( "DOI:10.5114/pdia.2013.33376") Annotation( "https://www.sciencedirect.com/topics/biochemistry-genetics-and-molecular-biology/melanocyte") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Melanocytes of the skin are specialized pigment cells crucial for producing and distributing melanin, the pigment determining skin, in addition to hair and eye color. Located in the basal layer of the epidermis, melanocytes serve as a natural sunscreen, protecting skin cells from harmful UV radiation. Structurally dendritic, they synthesize melanin through melanogenesis, transferring melanosomes to keratinocytes for UV shielding Beyond pigmentation and UV protection, melanocytes play roles in immune and inflammatory responses. They respond to environmental changes, communicate with skin cell types, and secrete cytokines and growth factors influencing skin homeostasis. Dysregulation may lead to pigment disorders like vitiligo and melasma or contribute to skin cancer, particularly malignant melanoma.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000458") -AnnotationAssertion(Annotation( "DOI:10.1101/cshperspect.a030510") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK279291") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/smooth-muscle-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1101/cshperspect.a030510") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK279291") Annotation( "https://www.sciencedirect.com/topics/immunology-and-microbiology/smooth-muscle-cell") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Smooth muscle cells of the prostate constitute a critical component of the prostatic stroma, enveloping glandular structures to support functions like secretion and fluid expulsion. Like other smooth muscles, smooth muscle cells of the prostate lack the striations characteristic of skeletal and cardiac muscle cell. Functionally, they play a crucial role in reproductive and urinary functions. Smooth muscle cells of the prostate contribute to the transport and expulsion of prostatic secretions into the urethra and respond dynamically to signaling molecules such as hormones and neurotransmitters. For instance, under sympathetic nervous stimulation, they contract and apply pressure on the gland's ducts, facilitating the secretion process.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000487") -AnnotationAssertion(Annotation( "DOI:10.1016/j.jcmgh.2015.05.005") Annotation( "DOI:10.1016/j.jhep.2012.10.011") Annotation( "DOI:10.1038/s41575-019-0125-y") Annotation( "DOI:10.1152/ajpgi.00227.2012") Annotation( "DOI:10.5009/gnl16033") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.jcmgh.2015.05.005") Annotation( "DOI:10.1016/j.jhep.2012.10.011") Annotation( "DOI:10.1038/s41575-019-0125-y") Annotation( "DOI:10.1152/ajpgi.00227.2012") Annotation( "DOI:10.5009/gnl16033") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Cholangiocytes, also known as biliary epithelial cells, are specialized epithelial cells that line the biliary tract, which constitutes the gall bladder and bile ducts inside the liver. Crucial to the maintenance of the liver's health and function, cholangiocytes have a key role in the modification and secretion of bile, a fluid produced by hepatocytes that is essential to digestion and the absorption of fats and vitamins. Cholangiocytes accomplish their primary function through the expression of a variety of transport proteins located on their apical and basolateral membranes, which propel bile acids and other contents of the bile into the biliary lumen. The hepatic bile, once secreted by the hepatocytes, is further modified by cholangiocytes via secretion and absorption processes. These processes help in the regulation of bile volume and composition, which is fundamental in ensuring the efficient digestion of dietary fats and fat-soluble vitamins and the excretion of cholesterol. In addition to their role in bile modification, cholangiocytes also perform several other integral functions. For instance, these cells express Toll-like receptors (TLRs) which allow cholangiocytes to initiate an immune response against pathogens in the biliary lumen. When functioning normally, these cells contribute to biliary integrity, hepatic architecture, and overall hepatic physiology. However, when they become pathological, they are involved in the progression of liver diseases, such as primary biliary cirrhosis and cholangiocarcinoma – the malignancy of the biliary tract.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000488") -AnnotationAssertion(Annotation( "DOI:10.1016/j.ccm.2021.08.005") Annotation( "DOI:10.1016/j.coi.2020.04.005") Annotation( "DOI:10.1152/ajplung.90587.2008") Annotation( "DOI:10.3389/fphys.2014.00284") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.ccm.2021.08.005") Annotation( "DOI:10.1016/j.coi.2020.04.005") Annotation( "DOI:10.1152/ajplung.90587.2008") Annotation( "DOI:10.3389/fphys.2014.00284") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Mesothelial cells of the pleura form a significant part of the pleural membrane, a thin, double-layered serous membrane that lines the thoracic cavity and encompasses the lungs. These specialized cells contribute to the pleura's key function of producing a lubricating serous fluid, which reduces friction between the lung's outer surface (visceral pleura) and the inner lining of the thoracic cavity (parietal pleura) during respiration. The cellular structure of mesothelial cells, characterized by microvilli on their surface, aids in the secretion and absorption of the pleural fluid, effectively supporting the smooth expansion and contraction of the lungs. Dysregulation in mesothelial cells can lead to pathologies, including pleural effusion and malignant mesothelioma. Mesothelial cells of the pleura display unique immunologic properties. They act as a first line of defense against infection because they are able to recognize pathogens and respond by secreting various cytokines and chemokines. Additionally, these cells are directly involved in the translocation of immune cells into the pleural cavity during inflammatory response, thereby playing an active role in the immune response within the pleural environment. In the event of pleural injury, these cells are also involved in the mesothelial-mesenchymal transition, a process that allows mesothelial cells to transdifferentiate into myofibroblasts and promote tissue repair.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000491") -AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(03)00242-5") Annotation( "DOI:10.1152/physrev.00026.2003") Annotation( "DOI:10.1371/journal.pone.0276978") Annotation( "DOI:10.3389/fphys.2014.00221") Annotation( "DOI:10.3390/jdb7020007") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/S1357-2725(03)00242-5") Annotation( "DOI:10.1152/physrev.00026.2003") Annotation( "DOI:10.1371/journal.pone.0276978") Annotation( "DOI:10.3389/fphys.2014.00221") Annotation( "DOI:10.3390/jdb7020007") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Mesothelial cells of visceral pleura are specialized epithelial cells that line the inner layer of the pleura, the membrane that envelops the lungs. Positioned adjacent to the lung tissue, these cells form a protective barrier and contribute to the structure of the visceral pleura. They are characterized by their cuboidal to squamous epithelial shape and the presence of microvilli on their surface, a feature aiding in fluid and solute exchange between the pleura and the lungs. The primary function of mesothelial cells of visceral pleura is to secrete a lubricating serous fluid to facilitate smooth, frictionless lung movement within the thoracic cavity during respiration. This helps in the prevention of trauma or damage stemming from the constant rubbing of the lung tissue against the chest wall, hence playing a pivotal role in maintaining respiratory function. Besides fluid secretion, these cells have an essential role in the transportation of fluids and particles across the pleura, as well as in inflammation, wound healing, and tissue repair processes within the pleura. Mesothelial cells of visceral pleura are notably implicated in the development of pleural diseases such as pleural effusion and pleural mesothelioma, a rare and aggressive form of cancer primarily linked with exposure to asbestos. Alterations, such as hyperplasia or metaplasia, may occur in these mesothelial cells under pathological conditions.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000493") -AnnotationAssertion(Annotation( "DOI:10.1016/bs.ircmb.2022.01.005") Annotation( "DOI:10.1172/jci.insight.161078") Annotation( "DOI:10.3389/fimmu.2021.681748") Annotation( "DOI:10.3389/fphys.2017.00837") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/bs.ircmb.2022.01.005") Annotation( "DOI:10.1172/jci.insight.161078") Annotation( "DOI:10.3389/fimmu.2021.681748") Annotation( "DOI:10.3389/fphys.2017.00837") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney resident macrophages are a heterogeneous population of immune cell found in the cortex and medullary regions of the kidney as well as within and surround the glomeruli. Their primary role involves maintaining homeostasis and immune surveillance in the kidney microenvironment, defending against ascending urinary infections. Beyond traditional macrophage roles such as phagocytosis and antigen presentation, kidney resident macrophages also contribute significantly to the response to kidney injury including tissue repair and angiogenesis. Under normal physiological conditions, kidney resident macrophages work to maintain the balance of the kidney's microenvironment by removing dead cells, pathogens and cellular debris, to limit inflammation and tissue damage. They are ‘professional’ phagocytes, clearing pathogens and debris by engulfing and digesting these harmful materials. Moreover, they are capable of presenting antigens to other immune cells, which aids in the activation of the adaptive immune response. Kidney resident macrophages show a high degree of plasticity: in response to environmental stimuli they change their morphology and cell surface markers. For example, in response to acute injury, some macrophages adopt a pro-inflammatory phenotype and augment tissue damage; once the injury stimulus is removed, they may change to mediate tissue repair. While kidney macrophages contribute to wound healing, tissue repair and regeneration by producing key growth factors and cytokines that stimulate cell proliferation, collagen production, and blood vessel formation, dysregulation in their activity can also lead to progressive inflammation and fibrosis, common features in chronic kidney disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000698") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c110052") Annotation( "DOI:10.1007/s00418-021-02033-5") Annotation( "DOI:10.1097/MNH.0b013e32820ac850") Annotation( "DOI:10.1681/ASN.2019040415") Annotation( "DOI:10.23876/j.krcp.2017.36.4.305") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c110052") Annotation( "DOI:10.1007/s00418-021-02033-5") Annotation( "DOI:10.1097/MNH.0b013e32820ac850") Annotation( "DOI:10.1681/ASN.2019040415") Annotation( "DOI:10.23876/j.krcp.2017.36.4.305") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney connecting tubule epithelial cells are found in collecting duct system of the kidney, specifically in at the interconnection segment between the nephron and the collecting ducts. The epithelial cells lining the connecting tubule are crucial for the journey of filtrate before it empties into urine-collecting ducts and therefore play a critical role in maintaining the body’s fluid and electrolyte balance. One of the principal functions of connecting tubule epithelial cells involves the selective reabsorption and secretion of various solutes. This reabsorption process includes critical regulatory electrolytes such as sodium ions (Na+), chloride ions (Cl-), and bicarbonate ions (HCO3-), thus playing a role in the regulation of the acid-base homeostasis. Their function also involves the secretion of potassium ions (K+) into the renal tubular fluid. The connecting tubule epithelial cells are primarily intercalated cells, which can be categorized into two types according to their specific functions: type A intercalated cells, which secrete acid, and the type B intercalated cells, which secrete base (bicarbonate) and reabsorb chloride. By regulating the concentrations of these ions, as well as reabsorption of water, connecting tubule epithelial cells contribute substantively to maintaining the body's electrolyte levels, pH balance, and overall homeostasis. In addition to intercalating cells, the collecting duct system also has principal cells, which are characterized by the expression of the water channel AQP2. AQP2-expressing cells have also been identified in connecting tubules, but there is some controversy whether these represent a unique cell type as there are some morphologic differences compared with principal cells.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000768") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c110060") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1159/000092212") Annotation( "DOI:10.1681/ASN.2019040415") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c110060") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1159/000092212") Annotation( "DOI:10.1681/ASN.2019040415") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney proximal convoluted tubule epithelial cells line the most voluminous part of the kidney's nephron, the proximal convoluted tubule, located in the cortex of the kidney. These cells are primarily involved in reabsorption and secretion processes, key functions that facilitate homeostasis within the body. Proximal convoluted tubule epithelial cells extensively function in the reabsorption process to a greater degree than their counterparts in the proximal straight tubule. Approximately two-thirds of filtrated sodium and water, nearly all nutrient sugars and amino acids, and a significant portion of bicarbonates pass through the proximal convoluted tubule, are effectively absorbed by the epithelial cells, and reenter the bodily circulation. By absorbing bicarbonates and secreting protons into the tubular fluid, proximal convoluted tubule epithelial cells also contribute to the pH regulation in the body . In addition to reabsorption, proximal convoluted tubule epithelial cells also play a critical role in the excretion of waste metabolites and xenobiotics. These cells possess a range of transporters that secrete metabolic end products and certain drugs, hence aiding in their elimination. This function underlines their importance for body detoxification.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000838") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c110061") Annotation( "DOI:10.1038/nrd4461") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1681/ASN.2019040415") Annotation( "DOI:10.2215/CJN.10391012") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c110061") Annotation( "DOI:10.1038/nrd4461") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1681/ASN.2019040415") Annotation( "DOI:10.2215/CJN.10391012") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney proximal straight tubule epithelial cells line the straight portion of the proximal tubule of the nephron, extending from the cortical medullary ray into the outer stripe of the outer medulla. They are characterized by a brush border of dense microvilli on their apical surface, enhancing the cell's surface area and aiding reabsorption, though reabsorption occurs more heavily in the proximal convoluted tubule preceding the straight portion of the tubule. The primary function of the kidney proximal straight tubule epithelial cells is to reabsorb the filtrate that the kidney produces, which includes glucose, sodium, and water. The reabsorption occurs through both passive and active transport methods. These cells exhibit enzyme-rich microvilli that also absorb small proteins and peptides. Consequently, they are involved in the regulation of plasma levels of various substances, including glucose, amino acids, and electrolytes, and maintain the body's pH homeostasis by secreting H+ ions into the urine and absorbing bicarbonate ions. These cells are also reported to be involved in drug metabolism. Many drugs are filtered from the blood by the kidneys and subsequently reabsorbed by the kidney proximal tubule epithelial cells back into the bloodstream. Furthermore, these cells play a significant role in the progression of kidney diseases. For example, in diabetic nephropathy and acute kidney injury, kidney proximal straight tubule epithelial cells can undergo cellular changes and result in malfunctions such as reduced reabsorption and increased excretion.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000839") -AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c140002") Annotation( "DOI:10.1007/s40620-021-01032-y") Annotation( "DOI:10.1152/physrev.2000.80.1.277") Annotation( "DOI:10.1681/ASN.2019040415") Annotation( "DOI:10.2215/CJN.05920613") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/cphy.c140002") Annotation( "DOI:10.1007/s40620-021-01032-y") Annotation( "DOI:10.1152/physrev.2000.80.1.277") Annotation( "DOI:10.1681/ASN.2019040415") Annotation( "DOI:10.2215/CJN.05920613") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney distal convoluted tubule epithelial cells, often referred to simply as distal convoluted tubule (DCT) cells, constitute a significant portion of the nephron -- the functional building unit of the kidney involved in filtration and reabsorption of substances from blood. The DCT epithelial cells form lining of the distal convoluted tubule, a segment of the nephron located after the loop of Henle and before the collecting tubule. These cells are characterized by unique morphological features such as small size, low height and fewer microvilli compared to proximal convoluted tubule epithelial cells. These cells perform a host of vital functions contributing to the regulation of extracellular fluid volume and electrolyte balance, blood pressure and pH. While the DCT is less prominent in water and sodium reabsorption compared to the proximal tubule and the loop of Henle, it plays a crucial role in fine-tuning the process. DCT epithelial cells are responsible for the final adjustments of sodium, potassium and calcium reabsorption, thereby affecting overall fluid and electrolyte balance, blood pressure regulation and bone health. The reabsorption process is regulated by hormones including aldosterone and parathyroid hormone. The distal convoluted tubule (DCT) also plays a significant role in acid-base homeostasis by secreting protons (H+) into the tubular fluid and reabsorbing bicarbonate ions (HCO3-), thus regulating the pH of the blood. The ability of DCT cells to adjust urine concentration plays a part in maintaining the body’s pH balance and preventing conditions like acidosis or alkalosis. In summary, kidney distal convoluted tubule epithelial cells perform functions that are crucial to maintaining body's homeostasis.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000849") -AnnotationAssertion(Annotation( "DOI:10.1073/pnas.0736323100") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1161/HYPERTENSIONAHA.115.04739") Annotation( "DOI:10.1681/ASN.2009070759") Annotation( "DOI:10.1681/ASN.2015050515") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1073/pnas.0736323100") Annotation( "DOI:10.1146/annurev-physiol-052521-121841") Annotation( "DOI:10.1161/HYPERTENSIONAHA.115.04739") Annotation( "DOI:10.1681/ASN.2009070759") Annotation( "DOI:10.1681/ASN.2015050515") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Macula densa epithelial cells, as part of the complex nephron structure, play a crucial role in kidney function. These cells, which make up the macula densa, help regulate blood pressure and the filtration rate of the glomerulus by sensing sodium chloride amounts in the kidney tubules. Found where the thick ascending limb of the Loop of Henle touches the afferent arteriole, macula densa epithelial cells are positioned in an optimal location to monitor and respond to changes in the filtrate composition. These cells maintain intraglomerular homeostasis. When the sodium chloride concentration is high, macula densa cells respond by secreting adenosine and vasoconstricting the afferent arteriole to decrease the glomerular filtration rate. Conversely, if the sodium chloride concentration in the filtrate is low, the production and release of nitric oxide cause vasodilation to enhance the glomerular filtration rate. Macula densa epithelial cells also have a role in tubuloglomerular feedback mechanisms, an essential autoregulatory renal response. They send feedback to the glomerular mesangial cells, allowing constriction or relaxation of the afferent arteriole as needed depending on the content of the filtrate.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1000850") -AnnotationAssertion(Annotation( "DOI:10.1016/j.biocel.2010.05.015") Annotation( "DOI:10.1016/j.yexcr.2012.02.032") Annotation( "DOI:10.1111/j.1523-1755.2005.00260.x") Annotation( "DOI:10.2337/diacare.28.1.164") Annotation( "DOI:10.3389/fphys.2021.689083") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.biocel.2010.05.015") Annotation( "DOI:10.1016/j.yexcr.2012.02.032") Annotation( "DOI:10.1111/j.1523-1755.2005.00260.x") Annotation( "DOI:10.2337/diacare.28.1.164") Annotation( "DOI:10.3389/fphys.2021.689083") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The glomerular capillary endothelial cell comprises an intrinsic component of the glomerulus in the kidney. Glomeruli contain a network of capillaries where the first step of blood filtration takes place, with glomerular capillary endothelial cells acting as an integral part of this process. Unlike regular endothelial cells that line the vasculature, unique fenestrations (openings) characterize these cells, allowing for enhanced permeability and filtration efficacy. Together with the glomerular basement membrane and podocytes, the glomerular endothelial cells form the glomerular filtration barrier, which is responsible for blood filtration and therefore critical for removal of waste products, such as urea and creatinine, and excess substances, such as glucose and ions, from the bloodstream. The glomerular capillary endothelial cells' fenestrations permit the free flow of a variety of particles, barring larger, negatively charged proteins like serum albumin, enabling the formation of an ultrafiltrate. This ultrafiltrate is the primitive form of urine, which then passes through the proximal tubule for further processing and ultimately helps maintain systemic fluid and electrolyte balance. Moreover, the glomerular capillary endothelial cells are also believed to play a crucial role in renal pathologies. Any compromise to their structural integrity or functional performance can lead to kidney diseases, including but not limited to, diabetic nephropathy and glomerulonephritis. For instance, in diabetes, persistent hyperglycemia can injure these cells, leading to a compromised glomerular filtration barrier and proteinuria, indicating the loss of proteins in the urine.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001005") -AnnotationAssertion(Annotation( "DOI:10.1007/s00424-012-1126-7") Annotation( "DOI:10.1152/ajpregu.00332.2006") Annotation( "DOI:10.1152/physrev.00042.2012") Annotation( "DOI:10.2174/15701611113116660149") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00424-012-1126-7") Annotation( "DOI:10.1152/ajpregu.00332.2006") Annotation( "DOI:10.1152/physrev.00042.2012") Annotation( "DOI:10.2174/15701611113116660149") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney afferent arteriole cells are specialized cell types found in the renal vasculature, specifically in the afferent arterioles which help in maintaining the renal blood flow and glomerular filtration rate. They are essential for the proper functioning of the kidneys, contributing to the regulation of blood pressure and fluid balance within the organism. Located before the glomerulus, a tiny, intricate network of capillaries within the nephron, these cells help control the expansion or contraction of the arteriole, thus regulating the pressure and flow of blood into the glomerulus. Functionally, kidney afferent arteriole cells are largely involved in responding to changes in blood volume and systemic blood pressure. They achieve this by modulating the diameter of the afferent arteriole through a process known as autoregulation. Autoregulation involves two primary mechanisms: the myogenic response and the tubuloglomerular feedback. The myogenic response is essentially the intrinsic ability of the vascular smooth muscle to respond to pressure changes, while the tubuloglomerular feedback is the process where the macula densa cells located near the glomerulus respond to changes in fluid delivery rates in the tubules, sending signals to afferent arterioles to constrict or dilate accordingly. These cells also have significant interaction with renin-containing juxtaglomerular cells, which are located in the wall of afferent arterioles. Juxtaglomerular cells secrete the enzyme renin in response to low blood pressure or sympathetic nerve activity, initiating the renin-angiotensin-aldosterone system cascade that ultimately helps retain sodium and water to increase blood volume and pressure.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001006") -AnnotationAssertion(Annotation( "DOI:10.1152/ajpregu.2000.279.2.R629") Annotation( "DOI:10.1152/physrev.00042.2012") Annotation( "DOI:10.1159/000072054") Annotation( "DOI:10.1161/01.HYP.29.1.222") Annotation( "DOI:10.3748/wjg.v12.i34.5429") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1152/ajpregu.2000.279.2.R629") Annotation( "DOI:10.1152/physrev.00042.2012") Annotation( "DOI:10.1159/000072054") Annotation( "DOI:10.1161/01.HYP.29.1.222") Annotation( "DOI:10.3748/wjg.v12.i34.5429") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney efferent arteriole cells are highly adaptable vascular cells that make up the efferent arterioles in the kidney. These units are integral components of the renal microcirculation system, conducting blood away from the glomeruli after filtration occurs. Efferent arterioles directly affect glomerular hydrostatic pressure and, subsequently, the rate of glomerular filtration. They are the downstream channels that ensure the effective transportation of blood to the peritubular capillaries and vasa recta, critical for maintaining fluid balance and electrolyte homeostasis. Kidney efferent arteriole cells are exceptional in their ability to perform autoregulation—a critical dynamic control of intrarenal blood flow ensuring stable glomerular filtration rates. This is primarily achieved via myogenic and tubuloglomerular feedback mechanisms, where the cells respond to changes in pressure and the concentration of sodium chloride in the tubular fluid, and constrict or dilate depending on the detected changes. Cells associated with the efferent arteriole also respond to several systemic influences. Through complex intracellular signaling pathways, they respond to fluctuations in hormones (like angiotensin II and endothelin) and several other mediators such as nitric oxide and prostaglandins. Their adaptive capability helps maintain homeostasis and ensures the entire system's optimal performance under various physiological conditions. Dysfunctions in kidney efferent arteriole cells can lead to severe pathologies like hypertension and chronic kidney disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001009") -AnnotationAssertion(Annotation( "DOI:10.1007/s00360-018-1164-3") Annotation( "DOI:10.1146/annurev-physiol-021113-170350") Annotation( "DOI:10.1152/advan.00227.2022") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00360-018-1164-3") Annotation( "DOI:10.1146/annurev-physiol-021113-170350") Annotation( "DOI:10.1152/advan.00227.2022") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The kidney loop of Henle descending limb epithelial cells are highly specialized cells that play a crucial role in maintaining the body's salt and water balance. These cells line the descending limb of the loop of Henle, which is a portion of the renal tubule located in the kidneys, an organ responsible for filtering waste products, ions and extra water from the blood and converting them into urine. However, the descending limb epithelial cells are specifically involved in the countercurrent multiplication system, a mechanism to concentrate urine that involves both active and passive transport processes. These cells are notably permeable to water but relatively impermeable to solutes, a characteristic that dramatically influences the osmotic gradient in the renal medulla. As filtrate initially descends down the loop of Henle, water gets drawn out due to the increasing osmotic gradient, thereby concentrating the filtrate. Additionally, these epithelial cells are exceptionally thin walled, allowing water to freely diffuse across the membrane. Furthermore, the kidney loop of Henle descending limb epithelial cells are inextricably linked to the functioning of numerous diuretic drugs, which work by blocking the reabsorption of water and certain electrolytes in the kidneys, leading to increased urine production to rid the body of excess fluids.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001021") -AnnotationAssertion(Annotation( "DOI:10.1046/j.0001-6772.2003.01205.x") Annotation( "DOI:10.1093/ndt/gfl308") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1046/j.0001-6772.2003.01205.x") Annotation( "DOI:10.1093/ndt/gfl308") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney afferent arteriole endothelial cells are a specialized type of cell located within the kidneys, forming the inner lining of the afferent arterioles, which are responsible for delivering blood to the glomeruli - capillary networks responsible for filtration - from where the process of urine formation begins in the nephrons. The endothelial cells in the kidney afferent arterioles have a key function in regulating the blood flow and filtration. They have autocrine and paracrine signaling capabilities, meaning they can signal to themselves and other nearby cells. They produce nitric oxide, prostacyclin, and endothelin, which are powerful vasodilators and vasoconstrictors that regulate renal blood flow. These cells also engage in the mitigation of kidney injury and inflammation by promoting repair and regeneration, demonstrating the multifaceted roles these cells play in maintaining renal health.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001096") -AnnotationAssertion(Annotation( "DOI:10.1016/j.semcdb.2014.08.002") Annotation( "DOI:10.1016/j.semnephrol.2015.01.010") Annotation( "DOI:10.1038/s41581-021-00411-9") Annotation( "DOI:10.1681/ASN.2019111179") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.semcdb.2014.08.002") Annotation( "DOI:10.1016/j.semnephrol.2015.01.010") Annotation( "DOI:10.1038/s41581-021-00411-9") Annotation( "DOI:10.1681/ASN.2019111179") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Kidney efferent arteriole endothelial cells constitute a vital component of the kidney's microvascular system. They are unique endothelial cells found lining the walls of efferent arterioles, which transport blood away from the glomeruli in the kidney. The primary responsibilities of the kidney efferent arteriole endothelial cells involve controling renal blood flow, regulating glomerular filtration rate (GFR), and managing perfusion pressure. They do this by contracting and relaxing, effectively narrowing and widening the arteriole's lumen thereby controling the volume and rate of blood flow to the peritubular capillaries and creating the pressure gradient necessary for filtration in the glomerulus. Furthermore, kidney efferent arteriole endothelial cells show a high degree of plasticity in response to pathophysiological stimuli and can undergo structural and functional changes based on local needs. In conditions like hypertension and diabetes, these cells can experience hypertrophy and endothelial dysfunction, contributing to the progression of renal disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001099") -AnnotationAssertion(Annotation( "DOI:10.1016/B978-0-323-35515-5.00009-9") Annotation( "DOI:10.1053/j.ajkd.2005.01.008") Annotation( "DOI:10.1081/jdi-100101958") Annotation( "DOI:10.1111/apha.12026") Annotation( "DOI:10.1152/ajpregu.00657.2002") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/B978-0-323-35515-5.00009-9") Annotation( "DOI:10.1053/j.ajkd.2005.01.008") Annotation( "DOI:10.1081/jdi-100101958") Annotation( "DOI:10.1111/apha.12026") Annotation( "DOI:10.1152/ajpregu.00657.2002") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The vasa recta ascending limb cells are specialized epithelial cells that are part of the vasa recta in the kidney, a crucial component of the renal medulla that functions as a counter-current exchanger to maintain the concentration gradient required for water reabsorption. These particular cells are located in the ascending limb of the vasa recta, which takes blood flow from the medulla back to the cortex. The primary function of vasa recta ascending limb cells is to preserve the renal medulla's hypertonicity, necessary for the kidney's urine concentration mechanism. Unlike the vasa recta descending limb cells, the venous-like epithelial cells of the ascending vasa recta are highly fenestrated and lack pericyte coverage, which facilitates water reuptake. As the blood flows through the ascending limb, it loses solutes and gains water, which is driven by the high solute concentration in the surrounding interstitium. This process acts in concert with cellular action in the descending limb and contributions from the so-called Loop of Henle, which helps with water and sodium chloride retrieval from the urine. In the context of normal physiological processes, the function of the vasa recta ascending limb cells is essential in maintaining the body's overall fluid balance, electrolyte concentration, and systemic blood pressure. Any dysfunction in these cells may lead to impaired urine concentration or dilution capability of the kidney, potentially resulting in conditions like diabetes insipidus or hyponatremia.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001131") -AnnotationAssertion(Annotation( "DOI:10.1053/j.ajkd.2005.01.008") Annotation( "DOI:10.1081/jdi-100101958") Annotation( "DOI:10.1111/apha.12026#apha12026-bib-0039") Annotation( "DOI:10.1152/ajpregu.00657.2002") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1053/j.ajkd.2005.01.008") Annotation( "DOI:10.1081/jdi-100101958") Annotation( "DOI:10.1111/apha.12026#apha12026-bib-0039") Annotation( "DOI:10.1152/ajpregu.00657.2002") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Vasa recta descending limb cells are specialized epithelial cells found in the vasa recta, a network of blood vessels in the renal medulla. These cells line the interior surface of the descending limb of the vasa recta and play a crucial role in the filtering and regulation of substances within the blood. In contrast to vasa recta ascending limb cells, the arterial-like epithelial cells of the descending vasa recta are non-fenestrated and covered by a pericyte layer that regulates the medullary blood flow. They help keeping the body’s fluid and electrolyte balance in check through a process known as countercurrent exchange system, in which the cells of the descending limb are permeable to water but relatively impermeable to solutes, such as sodium and urea. As the blood descends into the medulla along its descending limb, water passively diffuses out of the vasa recta, concentrating the blood in solutes. In addition to their central role in water and solute exchange, these cells contribute to maintaining the medullary osmotic gradient, a critical function to concentrate urine. The osmotic gradient is created by the counterflow of water and solutes between the descending and ascending limbs of the vasa recta and the adjacent Loop of Henle. The selective permeability of the descending limb cells allows them to maintain this gradient, which in turn helps to conserve water, a vital role in the overall function of the renal system. Thus, vasa recta descending limb cells have a significant function in renal physiology, particularly in osmoregulation and fluid balance.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001285") -AnnotationAssertion(Annotation( "DOI:10.1038/nrurol.2011.144") Annotation( "DOI:10.1038/nrurol.2016.13") Annotation( "DOI:10.1038/s41579-020-0324-0") Annotation( "DOI:10.1038/s41585-020-0350-8") Annotation( "DOI:10.1080/03008880410015165") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nrurol.2011.144") Annotation( "DOI:10.1038/nrurol.2016.13") Annotation( "DOI:10.1038/s41579-020-0324-0") Annotation( "DOI:10.1038/s41585-020-0350-8") Annotation( "DOI:10.1080/03008880410015165") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Bladder urothelial cells form the urothelium in the bladder - a specific type of epithelial tissue that constitutes the inner lining of the bladder and other parts of the urinary tract including the renal pelvis, ureters and urethra. These unique cells are specialized to withstand the highly variable and sometimes harsh conditions present in the urinary system. Their key function is maintaining the barrier integrity of the urinary tract even when exposed to high volumes of urine and its solutes, and they play a crucial role in protecting underlying tissues from damage and infection. The bladder urothelial cells exhibit a remarkable spectrum of phenotypic versatility and have an extraordinary capacity to stretch and retract based on the degree of bladder filling and emptying. In their relaxed state, these cells appear large and cuboidal, but upon stretching, they become thinner and exhibit a squamous cell-like appearance. This distinctive feature enables them to adjust to the changes in the urinary bladder volume while remaining impermeable to urinary solutes, thereby preventing a potential toxicity to the bladder wall and infiltration into the bloodstream. Apart from their barrier function, bladder urothelial cells also express a number of sensor molecules or respond to thermal, mechanical and chemical stimuli and can release signaling molecules, thereby playing a key role in sensory mechanisms that communicate vital information about the bladder's mechanical state to the nervous system. This communication helps regulate the micturition cycle and maintain urinary continence. Recent studies have also indicated that these cells may play a role in immune responses, highlighting their importance in bladder physiology. Altered proliferation and differentiation of bladder urothelial cells are implicated in various diseases and conditions such as bladder cancer, interstitial cystitis, and urinary tract infections.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001428") -AnnotationAssertion(Annotation( "DOI:10.1038/nrurol.2016.13") Annotation( "DOI:10.1038/s41385-022-00565-0") Annotation( "DOI:10.1152/physrev.00030.2012") Annotation( "DOI:10.1152/physrev.00041.2019") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nrurol.2016.13") Annotation( "DOI:10.1038/s41385-022-00565-0") Annotation( "DOI:10.1152/physrev.00030.2012") Annotation( "DOI:10.1152/physrev.00041.2019") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Urethra urothelial cells are highly specialized epithelial cells in the urethra, which is a tubelike structure that carries urine from the bladder to the external urethral orifice. Urothelial cells form the urothelium – a stratified, transitional epithelium lining the bladder, ureters, renal pelvis and urethra. These cells are highly adapted to their specific environment and maintain the crucial function of sustaining the urinary tract's core roles, primarily to transit and store urine without auto-corrosion or pathogen proliferation. Within the urethra urothelial cells are primarily found in proximal two-thirds of the urethra, while the distal third is lined by stratified squamous epithelial cells. The urothelial cells are renowned for their remarkable impermeability and ability to stretch and recoil during the various stages of urine filling and emptying. They form a tight barrier that prevents reabsorption of harmful byproducts and toxic substances present in the urine, guarding the deeper tissues and bloodstream against potential damage. This is largely attributed to specialized junctions, namely zonulae occludentes or tight junctions, in the umbrella cell layer (one of three layers of the urothelium) that seal the intercellular space between adjacent cells. Together with the urothelial plaque (an apical membrane plaque comprised of uroplakin proteins covering the umbrella cells at the luminal surface), the junctional complexes form a very effective permeability barrier that regulates the passage of water and ion from urine to the underlying tissue. A noteworthy feature of urothelial cells is their regulatory and sensory roles involving communication with the underlying layers about the filling and emptying status of the urinary tract. The cells also manage a fine balance between proliferation and differentiation, with the basal cells providing a ready pool of cells to replace the superficial layer when damaged. Their remarkable ability for regeneration and turnover, and their response to signals for repair underpins the durability and functionality of the urothelial tract.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001430") -AnnotationAssertion(Annotation( "DOI:10.1007/s11906-015-0538-0") Annotation( "DOI:10.1016/B978-0-12-386456-7.05402-2") Annotation( "DOI:10.1016/j.biocel.2022.106261") Annotation( "DOI:10.1073/pnas.1710964114") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s11906-015-0538-0") Annotation( "DOI:10.1016/B978-0-12-386456-7.05402-2") Annotation( "DOI:10.1016/j.biocel.2022.106261") Annotation( "DOI:10.1073/pnas.1710964114") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The kidney collecting duct principal cell is a highly specialized type of cell found in the late distal convoluted tubule and collecting duct of the kidney's nephron. Principal cells are located at the final segments of the renal tubules, where they play a pivotal role in key homeostatic processes. One of their fundamental functions of kidney collecting duct principal cells is the regulation of water reabsorption, which is mediated by aquaporins (water channel proteins). Antidiuretic hormones, such as vasopressin, can stimulate the redistribution of these water channels from an intracellular pools to the apical plasma membrane of the principal cell; translocation of aquaporin (specifically, AQP2) is associated with an increase of osmotic water permeability. The water reabsorption affects the concentration of the final urine; these cells are therefore directly involved in the maintenance of the body's fluid balance. Kidney collecting duct principal cells also participate in sodium and potassium ions regulation. They reabsorb sodium ions from the tubular fluid back into the bloodstream, a process facilitated by the action of aldosterone, a hormone released by the adrenal glands. Similarly, the principal cells secrete potassium ions into the tubular fluid in response to aldosterone, contributing to the regulation of potassium levels in the body. Impaired function of cells can lead to various renal diseases and disorders, highlighting the vital role of kidney collecting duct principal cells in the body's homeostatic processes.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001431") -AnnotationAssertion(Annotation( "DOI:10.1007/978-3-211-99390-3_106") Annotation( "DOI:10.1152/physiol.00008.2011") Annotation( "DOI:10.1152/physrev.00011.2019") Annotation( "DOI:10.2215/CJN.08880914") Annotation( "DOI:10.3390/diseases2020071") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/978-3-211-99390-3_106") Annotation( "DOI:10.1152/physiol.00008.2011") Annotation( "DOI:10.1152/physrev.00011.2019") Annotation( "DOI:10.2215/CJN.08880914") Annotation( "DOI:10.3390/diseases2020071") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The kidney collecting duct intercalated cell, otherwise referred to simply as the intercalated cell, is a specialized epithelial cell type primarily found in the kidney's collecting duct system. It is among the main cells that line this final component of the nephron, crucial in the physiological system that maintains acid-base homeostasis. This unique cell type comes in mainly three subtypes as per the localization of their proton pumps: type A, type B, and non-A, non-B cells. Each subtype plays different roles in regulating acid-base balance and electrolyte homeostasis in the body under varying physiological conditions. Type A intercalated cells reabsorb bicarbonate and secrete protons into urine, leading to the production of acidic urine. These cells are typically prevalent on the renal cortex and function particularly in states of metabolic acidosis or potassium depletion. On the other hand, type B intercalated cells secrete bicarbonate and reabsorb protons; thus, they are crucial in alkaline urine production and potassium conservation. These cells are essential to counter metabolic alkalosis or situations of potassium surplus. The non-A, non-B cells have the characteristic features of both type A and B cells, but play a prominent role under conditions of chronically elevated aldosterone and angiotensin II levels. Through their differentiated roles, intercalated cells also facilitate the reabsorption of sodium and chloride ions, and water to maintain osmolarity. These cells' activity is finely regulated by hormonal signals like aldosterone, angiotensin II, vasopressin, and the parathyroid hormone. Intercalated cell activity can undergo a significant transformation under pathological conditions, and their dysregulation is associated with diseases such as distal renal tubular acidosis, hypertension, and chronic kidney disease.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001432") -AnnotationAssertion(Annotation( "DOI:10.1016/j.coph.2013.09.008") Annotation( "DOI:10.1038/mi.2017.73") Annotation( "DOI:10.1038/s41574-019-0168-8") Annotation( "DOI:10.1111/j.1365-2613.2011.00767.x") Annotation( "DOI:10.1146/annurev-physiol-021115-105439") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.coph.2013.09.008") Annotation( "DOI:10.1038/mi.2017.73") Annotation( "DOI:10.1038/s41574-019-0168-8") Annotation( "DOI:10.1111/j.1365-2613.2011.00767.x") Annotation( "DOI:10.1146/annurev-physiol-021115-105439") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Intestinal enteroendocrine cells are a highly specialized cell type found within the gastrointestinal epithelium. They account for approximately 1% of the intestinal epithelial cells in what is known as the largest endocrine organ in the human body. Intestinal enteroendocrine cells exhibit an exceptional diversity, both in terms of their secretory products and their distribution along the intestines, varying from the small intestine to the colon. The primary function of intestinal enteroendocrine cells is to maintain gut physiology and homeostasis. They are known for their hormone production and are the source of numerous bioactive molecules including somatostatin, serotonin, cholecystokinin, glucose-dependent insulinotropic peptide, motilin, neurotensin, and others. These hormones and neuropeptides act in autocrine, paracrine, or endocrine signaling, impacting various functions including intestinal motility, secretion, nutrient absorption, and gut barrier functions. Intestinal enteroendocrine cells also serve as the body's frontline of defense, playing an integral role in surveillance against lumen-occurring hazards including pathogen attack. Enteroendocrine cells are divided into ‘closed type’ and ‘open type’. Closed type cells are located close to the basal membrane and do not reach the lumen of the gut. In contrast, the apical side of the open-type intestinal enteroendocrine cells is directly exposed to luminal contents and thus can recognize bacterial metabolites and dietary nutrients. In response, these cells can modulate the immune system responses and thereby function as a key communicator between the gut microbiota and the host.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001516") -AnnotationAssertion(Annotation( "DOI:10.1007/978-3-030-32300-4_26") Annotation( "DOI:10.1007/s00265-006-0178-0") Annotation( "DOI:10.1111/j.1439-0272.1992.tb02636.x") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK499854/") Annotation( "https://www.sciencedirect.com/topics/veterinary-science-and-veterinary-medicine/seminal-vesicle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/978-3-030-32300-4_26") Annotation( "DOI:10.1007/s00265-006-0178-0") Annotation( "DOI:10.1111/j.1439-0272.1992.tb02636.x") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK499854/") Annotation( "https://www.sciencedirect.com/topics/veterinary-science-and-veterinary-medicine/seminal-vesicle") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Seminal vesicle glandular cells belong to a specialized group of epithelial cells that form the internal lining of the seminal vesicles, a pair of male reproductive organs. These cells are located within the complex tubuloalveolar glands that make up the seminal vesicles and are known for their unique pseudostratified columnar epithelium structure. The primary function of these cells is the secretion of a variety of substances that ultimately constitute around 70% of the fluid volume of semen. Seminal cells produce a high-fructose fluid that serves as an energy source for the spermatozoa and promotes their motility. They also secrete other essential substances like proteins, enzymes, vitamin C, prostaglandins, and various other compounds. Collectively, these substances help in the nourishment, protection, and transportation of the spermatozoa throughout the male reproductive system and during the ejaculation process. Secondary to the production of seminal fluid, the seminal vesicle glandular cells also play a role in the contraction of the seminal vesicles during ejaculation. The contraction of these glands, induced by sympathetic nerves, ensures the efficient propulsion of the seminal fluid mixed with spermatozoa into the ejaculatory ducts and subsequently to the urethra. Notably, any malfunction or pathological condition affecting these cells can impact male fertility, emphasizing the importance of understanding the intricate functions of seminal vesicle glandular cells in maintaining the healthy physiological function of male reproduction.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001597") -AnnotationAssertion(Annotation( "DOI:10.1038/nbt.2247") Annotation( "DOI:10.1038/nrn1824") Annotation( "DOI:10.1177/0271678X156179") Annotation( "DOI:10.1186/s12987-020-00230-3") Annotation( "DOI:10.3389/fnins.2023.1047778") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/nbt.2247") Annotation( "DOI:10.1038/nrn1824") Annotation( "DOI:10.1177/0271678X156179") Annotation( "DOI:10.1186/s12987-020-00230-3") Annotation( "DOI:10.3389/fnins.2023.1047778") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Cerebral cortex endothelial cells are unique endothelial cells responsible for the formation and regulation of the blood-brain barrier (BBB), a specialized structure that separates the circulating blood from the cerebral neural tissue. These cells line the inner surface of nearly all blood vessels that irrigate the cerebral cortex, the outermost layer of the brain involved with complex cognitive functions such as thought, perception, and memory formation. Cerebral cortex endothelial cells are integral to maintaining brain health and function due to their role in regulating brain microenvironment and controlling the passage of essential molecules and cells. The endothelial cells in the cerebral cortex are distinctive due to their highly selective permeability, largely attributable to the existence of unique tight junctions which maintain the barrier function of the BBB. The tightly interconnected structures of these cells prevent most substances from freely diffusing into the brain tissue, ensuring that harmful agents such as toxins, pathogens, or peripheral immune cells don't reach the brain and compromise its function. These cells also express multiple transporters and enzymes, allowing the selective entry of necessary nutrients and metabolites while excluding toxic substances. Cerebral cortex endothelial cells also participate in neurovascular coupling, a process where localized neuronal activity increases cerebral blood flow to satisfy the local metabolic demands. The endothelial cells work in coordination with neurons and other cell types such as astrocytes and pericytes to mediate this process. They have been thought to regulate local blood flow by releasing substances in response to neuronal activity. Additionally, these cells are involved in many pathological conditions. Breakdown or dysfunction of the BBB often marked by altered endothelial cell function is linked to numerous neurological disorders including stroke, multiple sclerosis, Alzheimer’s disease, and brain tumors.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001602") -AnnotationAssertion(Annotation( "DOI:10.1002/JLB.3RU0720-418R") Annotation( "DOI:10.1016/j.immuni.2022.08.010") Annotation( "DOI:10.1189/jlb.70.2.163") Annotation( "DOI:10.3389/fimmu.2021.753940/full") Annotation( "DOI:10.3390/cells10040897") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/JLB.3RU0720-418R") Annotation( "DOI:10.1016/j.immuni.2022.08.010") Annotation( "DOI:10.1189/jlb.70.2.163") Annotation( "DOI:10.3389/fimmu.2021.753940/full") Annotation( "DOI:10.3390/cells10040897") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Lung macrophages refer to macrophage populations in the lungs, including the tissue-resident alveolar macrophages and interstitial macrophages, located in the air spaces and the connective tissue of the lung, respectively, and recruited macrophages. They are important in maintaining tissue homeostasis and immunity. The primary function of lung macrophages is to patrol and maintain the pulmonary system, where they serve as the first line of defense against airborne pathogens and inhaled particles. They perform phagocytosis of pathogens, particulate matter, dead cells, and other cellular debris from the lungs to maintain clear air passages and optimize gas exchange. They may also participate in tissue repair and the resolution of inflammation following injury. Lung macrophages also play a critical role in immune regulation. They help to initiate an immune response by secreting cytokines and other inflammatory mediators and by presenting antigens from the pathogens they have engulfed to induce T cell responses. However, if activated improperly, lung macrophages can contribute to inflammatory diseases of the lung such as chronic obstructive pulmonary disease (COPD) or asthma.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_1001603") -AnnotationAssertion(Annotation( "DOI:10.1002/rmb2.12088") Annotation( "DOI:10.1093/humupd/dmw004") Annotation( "DOI:10.1111/j.1749-6632.2010.05938.x") Annotation( "DOI:10.3390/ijms21114092") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/rmb2.12088") Annotation( "DOI:10.1093/humupd/dmw004") Annotation( "DOI:10.1111/j.1749-6632.2010.05938.x") Annotation( "DOI:10.3390/ijms21114092") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Decidual cells, originating from the endometrial lining in the uterus, undergo significant transformations during pregnancy and are integral to its preparation and maintenance. Their conversion from endometrial fibroblast to a secretory phenotype, known as decidualization, occurs as a normal process in the menstrual cycle or in response to embryo implantation. With crucial roles in regulating maternal immune responses and placental development, decidual cells have a twofold function: preventing rejection of the conceptus by the mother's immune system and guiding placental development to avoid excessive trophoblast invasion leading to pre-eclampsia. During decidualization, these cells secrete proteins and growth factors supporting embryo implantation and sustaining early pregnancy. Furthermore, decidual cells modulate the activities of natural killer cells, macrophages, and T cells in the uterus, maintaining a balance of pro-inflammatory and anti-inflammatory conditions during pregnancy.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_2000002") -AnnotationAssertion(Annotation( "DOI:10.1038/s41423-023-01042-9") Annotation( "DOI:10.1038/s41467-019-14127-9") Annotation( "DOI:10.3389/fimmu.2023.1235812") Annotation( "DOI:10.3389/fphys.2020.00509") Annotation( "DOI:10.3389/fphys.2020.577584") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1038/s41423-023-01042-9") Annotation( "DOI:10.1038/s41467-019-14127-9") Annotation( "DOI:10.3389/fimmu.2023.1235812") Annotation( "DOI:10.3389/fphys.2020.00509") Annotation( "DOI:10.3389/fphys.2020.577584") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. A dermis lymphatic vessel endothelial cell is a specialized type of cell that lines the interior surface of lymphatic vessels present in the dermal layer of the skin. These cells are organized into a single layer, forming an integral part of the lymphatic system which acts as the body's secondary circulatory system. The primary function of dermis lymphatic vessel endothelial cells is to facilitate the transportation and regulation of the lymph fluid within the dermis. They act as a barrier, contributing to fluid homeostasis, lipid transport, and immune cell trafficking. These cells are uniquely equipped to let fluid and larger molecules, including proteins and immune cells, into the lymphatic vessels from the surrounding interstitial space. This is enabled by the specialized junctions between the endothelial cells and the presence of primary lymphatic valves that prevent the backflow of lymph. Another paramount role of dermis lymphatic vessel endothelial cells is their involvement in immune response. By interacting with white blood cells, especially T-cells and dendritic cells, they can mediate local immune surveillance and contribute to both innate and adaptive immune responses. These cells are also implicated in a variety of pathological conditions, including lymphedema and metastasis in cancer.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_2000011") -AnnotationAssertion(Annotation( "DOI:10.1016/j.jid.2016.10.017") Annotation( "DOI:10.1080/19381980.2017.1375636") Annotation( "DOI:10.4161/derm.1.2.8472") Annotation( "https://doi.org/10.2147/CLEP.S323744") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/sebum") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.jid.2016.10.017") Annotation( "DOI:10.1080/19381980.2017.1375636") Annotation( "DOI:10.4161/derm.1.2.8472") Annotation( "https://doi.org/10.2147/CLEP.S323744") Annotation( "https://www.sciencedirect.com/topics/agricultural-and-biological-sciences/sebum") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Sebaceous gland cells, also known as sebocytes, are small, oil-producing glands present in the skin of mammals. These specialized cells are predominantly located in the dermis, particularly on the face, scalp, and upper body. The primary function of sebaceous gland cells is the production and secretion of sebum, a complex mixture of lipids including triglycerides, wax esters, squalene, and metabolites of fat-soluble vitamins (such as vitamin E), as well as free fatty acids. The production process involves a unique form of programmed cell death called holocrine secretion. In this process, as sebaceous gland cells mature, they progressively accumulate lipid droplets, before ultimately disintegrating and releasing their lipid-rich contents into the gland's ductal system. The sebum generated by these cells serves multiple purposes. It acts as a waterproofing agent, preventing excessive wetting or drying of the skin and averaging skin's water-holding capacity. Sebum also has antimicrobial properties, providing a natural defense barrier against certain bacterial and fungal species. Additionally, it contributes to the skin’s suppleness and prevents it from becoming brittle. An overproduction or imbalance of sebaceous gland cells can lead to common skin disorders, such as acne, seborrhea, or rosacea, illustrating their importance in skin health.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_2000021") -AnnotationAssertion(Annotation( "DOI:10.1016/j.peptides.2018.05.012") Annotation( "DOI:10.1126/science.aam5894") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK535355") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK572070/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1016/j.peptides.2018.05.012") Annotation( "DOI:10.1126/science.aam5894") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK535355") Annotation( "https://www.ncbi.nlm.nih.gov/books/NBK572070/") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Ventricular cardiac muscle cells, or cardiomyocytes, are specialized cells found in the ventricular walls of the heart. They exhibit unique features, including centrally located nuclei, robust cytoskeletal structures, high mitochondria content, and intercalated discs, which contribute to their efficient pumping of blood throughout the body. The main role of ventricular cardiac muscle cells is to coordinate contractions, enabling the heart to pump blood throughout the body. This is achieved through controlled ion movement across cell membranes, generating rhythmic electrical signals called action potentials. Intercalated discs between these cells aid synchronized contractions by allowing efficient signal spread through gap junctions. The high mitochondrial content ensures a constant energy supply for this ongoing contraction cycle. Ventricular cardiac muscle cells contribute not only mechanically but also biochemically by secreting essential hormones like atrial natriuretic peptide and B-type natriuretic peptide for blood pressure regulation and cardiac remodeling. These cells exhibit low turnover under normal conditions, emphasizing their long-term stability, yet have limited regenerative capacity following cardiac injuries. Dysfunction in these cells can result in significant cardiac issues, including heart failure.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_2000046") -AnnotationAssertion(Annotation( "DOI:10.1007/s00018-019-03104-6") Annotation( "DOI:10.3389/fimmu.2020.00343") Annotation( "DOI:10.3390/nu15163564") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1007/s00018-019-03104-6") Annotation( "DOI:10.3389/fimmu.2020.00343") Annotation( "DOI:10.3390/nu15163564") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. The placental villous trophoblast, a highly specialized cell type crucial for placental development, plays a key role in facilitating exchanges between the maternal bloodstream and the developing fetus. Structurally, placental villous trophoblasts are situated on the villous tree structures, forming an outer epithelial layer. Comprising two subtypes—proliferating cytotrophoblasts forming the inner layer and terminally differentiated syncytiotrophoblasts constituting the outer layer in contact with maternal blood—these cells define the placental architecture. Functionally, placental villous trophoblasts play pivotal roles in forming and maintaining the placenta, ensuring the safe development of the fetus. Their primary function involves managing substance exchange between the mother and fetus, mediating the transfer of gases, nutrients, hormones, and waste materials to support optimal fetal growth. Additionally, these trophoblasts synthesize and release crucial hormones like human chorionic gonadotropin (hCG), which maintains the decidua and signals the mother's body to sustain pregnancy. Acting as a physical and immunological barrier, they protect the fetus from maternal immune cells and prevent the transmission of pathogens, contributing significantly to the success of pregnancy.") AnnotationAssertion( "https://cellxgene.cziscience.com/cellguide/CL_2000060") -AnnotationAssertion(Annotation( "DOI:10.1002/(SICI)1097-0029(19980415)41:2<98::AID-JEMT2>3.0.CO;2-M") Annotation( "DOI:10.1111/joa.13709") Annotation( "DOI:10.3389/fncel.2015.00480") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. +AnnotationAssertion(Annotation( "DOI:10.1002/(SICI)1097-0029(19980415)41:2<98::AID-JEMT2>3.0.CO;2-M") Annotation( "DOI:10.1111/joa.13709") Annotation( "DOI:10.3389/fncel.2015.00480") "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. Hypendymal cells are secretory cells located between the ependymal layer and the posterior commissure, forming the hypendmal layer of the subcommissural organ (SCO), a highly conserved gland that is part of the circumventricular system within the brain. Hypendymal cells are bipolar cells with a thin apical pole and basal process. Most of the ultrastructural characteristics of these cells are similar to those described for the ependymal cells (which are arranged into another layer – the ependyma). However, ependymal cells release their secretion into the ventricular cerebrospinal fluid whereas hypendymal cells project processes to the local blood vessels and to the subarachnoidal space.") diff --git a/src/patterns/dosdp-patterns/ExtendedDescription.yaml b/src/patterns/dosdp-patterns/ExtendedDescription.yaml index 08da07bd1..afdac2982 100644 --- a/src/patterns/dosdp-patterns/ExtendedDescription.yaml +++ b/src/patterns/dosdp-patterns/ExtendedDescription.yaml @@ -20,7 +20,7 @@ data_list_vars: annotations: - annotationProperty: description - text: "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies to only to some subtypes and species, and so should not be considered definitional. + text: "This extended description was generated by ChatGPT and reviewed by the CellGuide team, who added references, and by the CL editors, who approved it for inclusion in CL. It may contain information that applies only to some subtypes and species, and so should not be considered definitional. %s"