Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Elife ; 122023 Dec 21.
Article in English | MEDLINE | ID: mdl-38127424

ABSTRACT

Apoptosis and clearance of apoptotic cells via efferocytosis are evolutionarily conserved processes that drive tissue repair. However, the mechanisms by which recognition and clearance of apoptotic cells regulate repair are not fully understood. Here, we use single-cell RNA sequencing to provide a map of the cellular dynamics during early inflammation in mouse skin wounds. We find that apoptotic pathways and efferocytosis receptors are elevated in fibroblasts and immune cells, including resident Lyve1+ macrophages, during inflammation. Interestingly, human diabetic foot wounds upregulate mRNAs for efferocytosis pathway genes and display altered efferocytosis signaling via the receptor Axl and its ligand Gas6. During early inflammation in mouse wounds, we detect upregulation of Axl in dendritic cells and fibroblasts via TLR3-independent mechanisms. Inhibition studies in vivo in mice reveal that Axl signaling is required for wound repair but is dispensable for efferocytosis. By contrast, inhibition of another efferocytosis receptor, Timd4, in mouse wounds decreases efferocytosis and abrogates wound repair. These data highlight the distinct mechanisms by which apoptotic cell detection coordinates tissue repair and provides potential therapeutic targets for chronic wounds in diabetic patients.


Our skin is constantly exposed to potential damage from the outside world, and it is vital that any injuries are repaired quickly and effectively. Diabetes and many other health conditions can hamper wound healing, resulting in chronic wounds that are both painful and at risk of becoming infected, which can lead to serious illness and death of patients. After an injury to the skin, the wound becomes inflamed as immune cells rush to the site of injury to fight off infection and clear the wound of dead cells and debris. Some of these dead cells will have died by a highly controlled process known as apoptosis. These so-called apoptotic cells display signals on their surface that nearby healthy cells recognize. This triggers the healthy cells to eat the apoptotic cells to remove them from the wound. Previous studies have linked changes in cell death and the removal of dead cells to chronic wounds in patients with diabetes, but it remains unclear how removing dead cells from the wound affects healing. Justynski et al. used a genetic technique called single-cell RNA sequencing to study the patterns of gene activity in mouse skin cells shortly after a wound. The experiments found that, as the area around the wound started to become inflamed, the wounded cells produced signals of apoptosis that in turn triggered nearby healthy cells to remove them. Other signals relating to the removal of dead cells were also widespread in the mouse wounds and treating the wounds with drugs that inhibit these signals resulted in multiple defects in the healing process. Further experiments used the same approach to study samples of tissue taken from foot wounds in human patients with or without diabetes. This revealed that several genes involved in the removal of dead cells were more highly expressed in the wounds of diabetic patients than in the wounds of other individuals. These findings indicate that for wounds to heal properly it is crucial for the body to detect and clear apoptotic cells from the wound site. Further studies building on this work may help to explain why some diabetic patients suffer from chronic wounds and help to develop more effective treatments for them.


Subject(s)
Apoptosis , Efferocytosis , Humans , Animals , Mice , Apoptosis/genetics , Fibroblasts , Inflammation , Inhibition, Psychological
2.
Proc Natl Acad Sci U S A ; 120(37): e2306965120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669366

ABSTRACT

Fibrosis is regulated by interactions between immune and mesenchymal cells. However, the capacity of cell types to modulate human fibrosis pathology is poorly understood due to lack of a fully humanized model system. MISTRG6 mice were engineered by homologous mouse/human gene replacement to develop an immune system like humans when engrafted with human hematopoietic stem cells (HSCs). We utilized MISTRG6 mice to model scleroderma by transplantation of healthy or scleroderma skin from a patient with pansclerotic morphea to humanized mice engrafted with unmatched allogeneic HSC. We identified that scleroderma skin grafts contained both skin and bone marrow-derived human CD4 and CD8 T cells along with human endothelial cells and pericytes. Unlike healthy skin, fibroblasts in scleroderma skin were depleted and replaced by mouse fibroblasts. Furthermore, HSC engraftment alleviated multiple signatures of fibrosis, including expression of collagen and interferon genes, and proliferation and activation of human T cells. Fibrosis improvement correlated with reduced markers of T cell activation and expression of human IL-6 by mesenchymal cells. Mechanistic studies supported a model whereby IL-6 trans-signaling driven by CD4 T cell-derived soluble IL-6 receptor complexed with fibroblast-derived IL-6 promoted excess extracellular matrix gene expression. Thus, MISTRG6 mice transplanted with scleroderma skin demonstrated multiple fibrotic responses centered around human IL-6 signaling, which was improved by the presence of healthy bone marrow-derived immune cells. Our results highlight the importance of IL-6 trans-signaling in pathogenesis of scleroderma and the ability of healthy bone marrow-derived immune cells to mitigate disease.


Subject(s)
Basidiomycota , Scleroderma, Localized , Humans , Animals , Mice , Interleukin-6 , Endothelial Cells , Skin , Disease Models, Animal
3.
bioRxiv ; 2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36824808

ABSTRACT

Wound repair requires the coordination of multiple cell types including immune cells and tissue resident cells to coordinate healing and return of tissue function. Diabetic foot ulceration is a type of chronic wound that impacts over 4 million patients in the US and over 7 million worldwide (Edmonds et al., 2021). Yet, the cellular and molecular mechanisms that go awry in these wounds are not fully understood. Here, by profiling chronic foot ulcers from non-diabetic (NDFUs) and diabetic (DFUs) patients using single-cell RNA sequencing, we find that DFUs display transcription changes that implicate reduced keratinocyte differentiation, altered fibroblast function and lineages, and defects in macrophage metabolism, inflammation, and ECM production compared to NDFUs. Furthermore, analysis of cellular interactions reveals major alterations in several signaling pathways that are altered in DFUs. These data provide a view of the mechanisms by which diabetes alters healing of foot ulcers and may provide therapeutic avenues for DFU treatments.

4.
bioRxiv ; 2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36711968

ABSTRACT

Apoptosis and clearance of apoptotic cells via efferocytosis are evolutionarily conserved processes that drive tissue repair. However, the mechanisms by which recognition and clearance of apoptotic cells regulate repair are not fully understood. Here, we use single-cell RNA sequencing to provide a map of the cellular dynamics during early inflammation in mouse skin wounds. We find that apoptotic pathways and efferocytosis receptors are elevated in fibroblasts and immune cells, including resident Lyve1 + macrophages, during inflammation. Interestingly, human diabetic foot wounds upregulate mRNAs for apoptotic genes and display increased and altered efferocytosis signaling via the receptor Axl. During early inflammation in mouse wounds, we detect upregulation of Axl in dendritic cells and fibroblasts via TLR3-independent mechanisms. Inhibition studies in vivo in mice reveal that Axl signaling is required for wound repair but is dispensable for efferocytosis. By contrast, inhibition of another efferocytosis receptor, Timd4, in mouse wounds decreases efferocytosis and abrogates wound repair. These data highlight the distinct mechanisms by which apoptotic cell detection coordinates tissue repair and provides potential therapeutic targets for chronic wounds in diabetic patients.

5.
Cell ; 185(25): 4675-4677, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36493748

ABSTRACT

Wound healing in adult mammalian tissues generally involves scarring instead of tissue regeneration. A study in this issue of Cell reveals that after injury, reindeer antler skin regenerates by priming regenerative genes in wound fibroblasts instead of forming a scar through an inflammatory gene program.


Subject(s)
Reindeer , Animals , Wound Healing , Cicatrix/pathology , Skin/pathology , Fibroblasts/pathology
6.
Dev Cell ; 57(24): 2699-2713.e5, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36493773

ABSTRACT

Angiogenesis, the growth of new blood vessels from pre-existing vessels, occurs during development, injury repair, and tumorigenesis to deliver oxygen, immune cells, and nutrients to tissues. Defects in angiogenesis occur in cardiovascular and inflammatory diseases, and chronic, non-healing wounds, yet treatment options are limited. Here, we provide a map of the early angiogenic niche by analyzing single-cell RNA sequencing of mouse skin wound healing. Our data implicate Langerhans cells (LCs), phagocytic, skin-resident immune cells, in driving angiogenesis during skin repair. Using lineage-driven reportersw, three-dimensional (3D) microscopy, and mouse genetics, we show that LCs are situated at the endothelial cell leading edge in mouse skin wounds and are necessary for angiogenesis during repair. These data provide additional future avenues for the control of angiogenesis to treat disease and chronic wounds and extend the function of LCs beyond their canonical role in antigen presentation and T cell immunity.


Subject(s)
Langerhans Cells , Wound Healing , Mice , Animals , Skin/blood supply , Neovascularization, Physiologic
7.
Cell Rep ; 41(8): 111675, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417855

ABSTRACT

Many human diseases are caused by mutations in nuclear envelope (NE) proteins. How protein homeostasis and disease etiology are interconnected at the NE is poorly understood. Specifically, the identity of local ubiquitin ligases that facilitate ubiquitin-proteasome-dependent NE protein turnover is presently unknown. Here, we employ a short-lived, Lamin B receptor disease variant as a model substrate in a genetic screen to uncover key elements of NE protein turnover. We identify the ubiquitin-conjugating enzymes (E2s) Ube2G2 and Ube2D3, the membrane-resident ubiquitin ligases (E3s) RNF5 and HRD1, and the poorly understood protein TMEM33. RNF5, but not HRD1, requires TMEM33 both for efficient biosynthesis and function. Once synthesized, RNF5 responds dynamically to increased substrate levels at the NE by departing from the endoplasmic reticulum, where HRD1 remains confined. Thus, mammalian protein quality control machinery partitions between distinct cellular compartments to address locally changing substrate loads, establishing a robust cellular quality control system.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Animals , Humans , Ubiquitin-Protein Ligases/metabolism , Membrane Proteins/metabolism , Endoplasmic Reticulum/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Mammals/metabolism
8.
Curr Opin Genet Dev ; 76: 101968, 2022 10.
Article in English | MEDLINE | ID: mdl-35988318

ABSTRACT

Mammalian tissue repair forms a scar that fills the injured area with a fibrotic lesion, limiting tissue function. Adipocytes, lipid-filled cells, well-known for energy storage and endocrine functions, can reside adjacent to or within many tissues, and are emerging as critical regulators of tissue repair. In this review, the plasticity and function of adipocytes to tissue repair and fibrosis in four tissues: skin, heart, skeletal muscle, and mammary gland, will be discussed. The dynamic nature of adipocytes as they release bioactive products, lipids, and adipokines, and their ability to form contractile fibroblasts, is emerging as an essential regulator of wound healing and tumorigenesis in multiple tissues. Thus, modulation of adipocytes may provide therapeutic avenues for regenerative medicine and cancer.


Subject(s)
Adipocytes , Wound Healing , Adipokines/genetics , Animals , Fibrosis , Humans , Lipids , Mammals , Regenerative Medicine , Wound Healing/genetics
9.
Cell Stem Cell ; 29(2): 179-181, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35120615

ABSTRACT

Tissue repair in adult mammals lacks the regenerative ability of many tissues in other adult organisms like axolotl and newts. In this issue of Cell Stem Cell, Mascharak et al. use multi-omics approaches to identify an essential role for the transcription factor Trps1 in Yap-inhibited fibroblasts' tissue regenerative responses in murine skin.


Subject(s)
Cicatrix , Wound Healing , Animals , Cicatrix/pathology , Fibroblasts/pathology , Mammals , Mice , Skin/pathology , Transcription Factors , Wound Healing/physiology
10.
Nat Commun ; 13(1): 181, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013299

ABSTRACT

Diabetic foot ulceration (DFU) is a devastating complication of diabetes whose pathogenesis remains incompletely understood. Here, we profile 174,962 single cells from the foot, forearm, and peripheral blood mononuclear cells using single-cell RNA sequencing. Our analysis shows enrichment of a unique population of fibroblasts overexpressing MMP1, MMP3, MMP11, HIF1A, CHI3L1, and TNFAIP6 and increased M1 macrophage polarization in the DFU patients with healing wounds. Further, analysis of spatially separated samples from the same patient and spatial transcriptomics reveal preferential localization of these healing associated fibroblasts toward the wound bed as compared to the wound edge or unwounded skin. Spatial transcriptomics also validates our findings of higher abundance of M1 macrophages in healers and M2 macrophages in non-healers. Our analysis provides deep insights into the wound healing microenvironment, identifying cell types that could be critical in promoting DFU healing, and may inform novel therapeutic approaches for DFU treatment.


Subject(s)
Diabetes Mellitus/genetics , Diabetic Foot/genetics , Fibroblasts/metabolism , Macrophages/metabolism , Transcriptome , Wound Healing/genetics , Biomarkers/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Foot/metabolism , Diabetic Foot/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/pathology , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/pathology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 11/genetics , Matrix Metalloproteinase 11/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Single-Cell Analysis/methods , Skin/metabolism , Skin/pathology , Exome Sequencing
11.
J Invest Dermatol ; 142(6): 1597-1606.e9, 2022 06.
Article in English | MEDLINE | ID: mdl-34808238

ABSTRACT

Fibrosis is the life-threatening, excessive accumulation of the extracellular matrix and is sometimes associated with a loss of lipid-filled cells in the skin and other organs. Understanding the mechanisms of fibrosis and associated lipodystrophy and their reversal may reveal new targets for therapeutic intervention. In vivo genetic models are needed to identify key targets that induce recovery from established fibrosis. Wnt signaling is activated in animal and human fibrotic diseases across organs. Here, we developed a genetically inducible and reversible Wnt activation model and showed that it is sufficient to cause fibrotic dermal remodeling, including extracellular matrix expansion and shrinking of dermal adipocytes. Upon withdrawal from Wnt activation, Wnt-induced fibrotic remodeling was reversed in mouse skin-fully restoring skin architecture. Next, we demonstrated CD26/ DPP4 is a Wnt/ß-catenin-responsive gene and a functional mediator of fibrotic transformation. We provide genetic evidence that the Wnt/DPP4 axis is required to drive fibrotic dermal remodeling and is associated with human skin fibrosis severity. Remarkably, DPP4 inhibitors can be repurposed to accelerate recovery from established Wnt-induced fibrosis. Collectively, this study identifies Wnt/DPP4 axis as a key driver of extracellular matrix homeostasis and dermal fat loss, providing therapeutic avenues to manipulate the onset and reversal of tissue fibrosis.


Subject(s)
Dipeptidyl Peptidase 4 , Skin Diseases , Animals , Dipeptidyl Peptidase 4/genetics , Fibroblasts/metabolism , Fibrosis , Mice , Skin/pathology , Skin Diseases/genetics , Skin Diseases/pathology , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
12.
Cell ; 184(15): 3852-3872, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34297930

ABSTRACT

Fibroblasts are diverse mesenchymal cells that participate in tissue homeostasis and disease by producing complex extracellular matrix and creating signaling niches through biophysical and biochemical cues. Transcriptionally and functionally heterogeneous across and within organs, fibroblasts encode regional positional information and maintain distinct cellular progeny. We summarize their development, lineages, functions, and contributions to fibrosis in four fibroblast-rich organs: skin, lung, skeletal muscle, and heart. We propose that fibroblasts are uniquely poised for tissue repair by easily reentering the cell cycle and exhibiting a reversible plasticity in phenotype and cell fate. These properties, when activated aberrantly, drive fibrotic disorders in humans.


Subject(s)
Disease , Fibroblasts/metabolism , Health , Animals , Cell Lineage , Humans , Molecular Targeted Therapy , Signal Transduction
13.
J Invest Dermatol ; 141(7): 1615-1621.e1, 2021 07.
Article in English | MEDLINE | ID: mdl-34167718

ABSTRACT

The scientific process depends on social interactions: communication and dissemination of research findings, evaluation and discussion of scientific work, and collaboration with other scientists. Social media, and specifically, Twitter has accelerated the ability to accomplish these goals. We discuss the ways that Twitter is used by scientists and provide guidance on navigating the academic Twitter community.


Subject(s)
Biomedical Research/methods , Communication , Information Dissemination , Social Media , Dermatology/methods , Humans , Research Personnel
14.
Elife ; 102021 03 29.
Article in English | MEDLINE | ID: mdl-33779546

ABSTRACT

While the mechanisms by which chemical signals control cell fate have been well studied, the impact of mechanical inputs on cell fate decisions is not well understood. Here, using the well-defined system of keratinocyte differentiation in the skin, we examine whether and how direct force transmission to the nucleus regulates epidermal cell fate. Using a molecular biosensor, we find that tension on the nucleus through linker of nucleoskeleton and cytoskeleton (LINC) complexes requires integrin engagement in undifferentiated epidermal stem cells and is released during differentiation concomitant with decreased tension on A-type lamins. LINC complex ablation in mice reveals that LINC complexes are required to repress epidermal differentiation in vivo and in vitro and influence accessibility of epidermal differentiation genes, suggesting that force transduction from engaged integrins to the nucleus plays a role in maintaining keratinocyte progenitors. This work reveals a direct mechanotransduction pathway capable of relaying adhesion-specific signals to regulate cell fate.


Subject(s)
Epidermis/physiology , Mechanotransduction, Cellular/physiology , Nuclear Lamina/physiology , Plakins/genetics , Animals , Cell Differentiation , Female , Integrins/metabolism , Lamin Type A/metabolism , Mice , Plakins/metabolism
16.
Cell Stem Cell ; 26(6): 880-895.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32302523

ABSTRACT

Mature adipocytes store fatty acids and are a common component of tissue stroma. Adipocyte function in regulating bone marrow, skin, muscle, and mammary gland biology is emerging, but the role of adipocyte-derived lipids in tissue homeostasis and repair is poorly understood. Here, we identify an essential role for adipocyte lipolysis in regulating inflammation and repair after injury in skin. Genetic mouse studies revealed that dermal adipocytes are necessary to initiate inflammation after injury and promote subsequent repair. We find through histological, ultrastructural, lipidomic, and genetic experiments in mice that adipocytes adjacent to skin injury initiate lipid release necessary for macrophage inflammation. Tamoxifen-inducible genetic lineage tracing of mature adipocytes and single-cell RNA sequencing revealed that dermal adipocytes alter their fate and generate ECM-producing myofibroblasts within wounds. Thus, adipocytes regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.


Subject(s)
Lipolysis , Myofibroblasts , Adipocytes , Animals , Macrophages , Mice , Skin
17.
Cell ; 181(3): 492-494, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32234524

ABSTRACT

The 2020 Canada Gairdner International Award has been awarded to Elaine Fuchs for her discovery of the role of adult skin stem cells in homeostasis, wound repair, inflammation, and cancer. These insights have established a foundation for basic knowledge on how adult stem cells form, maintain, and repair tissues and have provided the groundwork for additional exploration and discovery of pathways in other stem cell systems.


Subject(s)
Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Skin/metabolism , Animals , Awards and Prizes , Canada , Epidermal Cells/metabolism , Female , History, 20th Century , History, 21st Century , Homeostasis/physiology , Humans , Neoplasms/metabolism , Wound Healing/physiology
18.
Soft Matter ; 16(13): 3325-3337, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32196025

ABSTRACT

Surface tension governed by differential adhesion can drive fluid particle mixtures to sort into separate regions, i.e., demix. Does the same phenomenon occur in confluent biological tissues? We begin to answer this question for epithelial monolayers with a combination of theory via a vertex model and experiments on keratinocyte monolayers. Vertex models are distinct from particle models in that the interactions between the cells are shape-based, as opposed to distance-dependent. We investigate whether a disparity in cell shape or size alone is sufficient to drive demixing in bidisperse vertex model fluid mixtures. Surprisingly, we observe that both types of bidisperse systems robustly mix on large lengthscales. On the other hand, shape disparity generates slight demixing over a few cell diameters, a phenomenon we term micro-demixing. This result can be understood by examining the differential energy barriers for neighbor exchanges (T1 transitions). Experiments with mixtures of wild-type and E-cadherin-deficient keratinocytes on a substrate are consistent with the predicted phenomenon of micro-demixing, which biology may exploit to create subtle patterning. The robustness of mixing at large scales, however, suggests that despite some differences in cell shape and size, progenitor cells can readily mix throughout a developing tissue until acquiring means of recognizing cells of different types.


Subject(s)
Cadherins/genetics , Cell Adhesion/drug effects , Keratinocytes/drug effects , Cadherins/chemistry , Cell Shape/drug effects , Cell Size/drug effects , Humans , Surface Properties
19.
J Invest Dermatol ; 140(9): 1698-1705.e1, 2020 09.
Article in English | MEDLINE | ID: mdl-32032578

ABSTRACT

Dermal white adipose tissue (dWAT) expansion is associated with important homeostatic and pathologic processes in skin. Even though mTOR/protein kinase B signaling is important for adipogenesis, the role of regulated development of DNA damage responses 1 (REDD1), a negative regulator of mTOR/protein kinase B, is poorly understood. Loss of REDD1 in mice resulted in reduction of body mass, total fat, size of gonadal white adipose tissue, and interscapular brown adipose tissue. Inguinal subcutaneous white adipose tissue and dWAT in REDD1 knockouts were expanded compared with wild type mice. Size and number of mature adipocytes in dWAT were also increased in adult REDD1 knockouts. This dWAT phenotype was established around postnatal day 18 and did not depend on the hair growth cycle. Numbers of adipocyte precursor cells were lower in REDD1 knockout skin. In vitro analysis revealed increased differentiation of skin-derived REDD1 knockout adipocyte precursor cells as indicated by higher lipid accumulation and increased adipogenic marker expression. 3T3L1 cells overexpressing REDD1 had decreased sensitivity to differentiation. Overall, our findings indicate that REDD1 silencing induced expansion of dWAT through hypertrophy and hyperplasia. This REDD1-dependent mechanism of adipogenesis could be used to preferentially target skin-associated adipose tissue for therapeutic purposes.


Subject(s)
Adipocytes/pathology , Adipogenesis/genetics , Dermis/metabolism , Subcutaneous Fat/pathology , Transcription Factors/metabolism , 3T3 Cells , Animals , Cell Differentiation/genetics , Dermis/cytology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Hair Follicle/growth & development , Humans , Hyperplasia/genetics , Hypertrophy/genetics , Hypertrophy/pathology , Male , Mice , Mice, Knockout , Signal Transduction/genetics , Subcutaneous Fat/cytology , Transcription Factors/genetics
20.
Dev Cell ; 51(3): 296-298, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31689385

ABSTRACT

Multiple stem cells maintain and repair tissues, yet how they communicate is not well understood. In this issue of Developmental Cell, Veniaminova et al. (2019) report that each sebaceous gland is maintained by local stem cells and that Notch signaling regulates multiple aspects of their function, revealing tissue homeostasis mechanisms.


Subject(s)
Receptors, Notch , Sebaceous Glands , Cell Differentiation , Homeostasis , Lifting , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...