Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2805: 187-201, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39008183

RESUMEN

Epidermal tissues are among the most striking examples of planar polarity. Insect bristles, fish scales, and mammalian fur are all uniformly oriented along an animal's body axis. The collective alignment of epidermal structures provides a valuable system to interrogate the signaling mechanisms that coordinate cellular behaviors at both local and tissue-levels. Here, we provide methods to analyze the planar organization of hair follicles within the mouse epidermis. Hair follicles are specified and bud into the underlying dermis during embryonic development. Shortly after, follicle cells dynamically rearrange to orient each follicle toward the anterior of the animal. When directional signaling is disrupted, hair follicles become misoriented. In this chapter, we describe how to create a spatial map of hair follicle orientations to reveal tissue-scale patterns in both embryonic and postnatal skin. Additionally, we provide a live imaging protocol that can be used to monitor cell movements in embryonic skin explants to reveal the cellular behaviors that polarize the hair follicle itself.


Asunto(s)
Polaridad Celular , Epidermis , Folículo Piloso , Animales , Ratones , Folículo Piloso/citología , Folículo Piloso/embriología , Polaridad Celular/fisiología , Epidermis/embriología , Epidermis/metabolismo , Células Epidérmicas/citología , Movimiento Celular
2.
Cell Death Dis ; 15(7): 508, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39019868

RESUMEN

Epidermal stem cells orchestrate epidermal renewal and timely wound repair through a tight regulation of self-renewal, proliferation, and differentiation. In culture, human epidermal stem cells generate a clonal type referred to as holoclone, which give rise to transient amplifying progenitors (meroclone and paraclone-forming cells) eventually generating terminally differentiated cells. Leveraging single-cell transcriptomic data, we explored the FOXM1-dependent biochemical signals controlling self-renewal and differentiation in epidermal stem cells aimed at improving regenerative medicine applications. We report that the expression of H1 linker histone subtypes decrease during serial cultivation. At clonal level we observed that H1B is the most expressed isoform, particularly in epidermal stem cells, as compared to transient amplifying progenitors. Indeed, its expression decreases in primary epithelial culture where stem cells are exhausted due to FOXM1 downregulation. Conversely, H1B expression increases when the stem cells compartment is sustained by enforced FOXM1 expression, both in primary epithelial cultures derived from healthy donors and JEB patient. Moreover, we demonstrated that FOXM1 binds the promotorial region of H1B, hence regulates its expression. We also show that H1B is bound to the promotorial region of differentiation-related genes and negatively regulates their expression in epidermal stem cells. We propose a novel mechanism wherein the H1B acts downstream of FOXM1, contributing to the fine interplay between self-renewal and differentiation in human epidermal stem cells. These findings further define the networks that sustain self-renewal along the previously identified YAP-FOXM1 axis.


Asunto(s)
Diferenciación Celular , Células Epidérmicas , Proteína Forkhead Box M1 , Histonas , Células Madre , Humanos , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/genética , Células Madre/metabolismo , Células Madre/citología , Células Epidérmicas/metabolismo , Células Epidérmicas/citología , Histonas/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Proliferación Celular , Epidermis/metabolismo , Células Cultivadas
3.
Int J Mol Sci ; 25(11)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38891823

RESUMEN

In the interfollicular epidermis, keratinocyte stem cells (KSC) generate a short-lived population of transit amplifying (TA) cells that undergo terminal differentiation after several cell divisions. Recently, we isolated and characterized a highly proliferative keratinocyte cell population, named "early" TA (ETA) cell, representing the first KSC progenitor with exclusive features. This work aims to evaluate epidermis, with a focus on KSC and ETA cells, during transition from infancy to childhood. Reconstructed human epidermis (RHE) generated from infant keratinocytes is more damaged by UV irradiation, as compared to RHE from young children. Moreover, the expression of several differentiation and barrier genes increases with age, while the expression of genes related to stemness is reduced from infancy to childhood. The proliferation rate of KSC and ETA cells is higher in cells derived from infants' skin samples than of those derived from young children, as well as the capacity of forming colonies is more pronounced in KSC derived from infants than from young children's skin samples. Finally, infants-KSC show the greatest regenerative capacity in skin equivalents, while young children ETA cells express higher levels of differentiation markers, as compared to infants-ETA. KSC and ETA cells undergo substantial changes during transition from infancy to childhood. The study presents a novel insight into pediatric skin, and sheds light on the correlation between age and structural maturation of the skin.


Asunto(s)
Diferenciación Celular , Queratinocitos , Células Madre , Humanos , Lactante , Células Madre/citología , Células Madre/metabolismo , Queratinocitos/metabolismo , Queratinocitos/citología , Preescolar , Proliferación Celular , Células Epidérmicas/metabolismo , Células Epidérmicas/citología , Niño , Piel/citología , Piel/metabolismo , Femenino , Masculino , Epidermis/metabolismo , Células Cultivadas
4.
Development ; 151(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38934416

RESUMEN

Transit-amplifying (TA) cells are progenitors that undergo an amplification phase followed by transition into an extinction phase. A long postulated epidermal TA progenitor with biphasic behavior has not yet been experimentally observed in vivo. Here, we identify such a TA population using clonal analysis of Aspm-CreER genetic cell-marking in mice, which uncovers contribution to both homeostasis and injury repair of adult skin. This TA population is more frequently dividing than a Dlx1-CreER-marked long-term self-renewing (e.g. stem cell) population. Newly developed generalized birth-death modeling of long-term lineage tracing data shows that both TA progenitors and stem cells display neutral competition, but only the stem cells display neutral drift. The quantitative evolution of a nascent TA cell and its direct descendants shows that TA progenitors indeed amplify the basal layer before transition and that the homeostatic TA population is mostly in extinction phase. This model will be broadly useful for analyzing progenitors whose behavior changes with their clone age. This work identifies a long-missing class of non-self-renewing biphasic epidermal TA progenitors and has broad implications for understanding tissue renewal mechanisms.


Asunto(s)
Células Epidérmicas , Epidermis , Células Madre , Animales , Ratones , Células Madre/citología , Células Madre/metabolismo , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Epidermis/metabolismo , Proliferación Celular , Linaje de la Célula , Homeostasis , Diferenciación Celular , Autorrenovación de las Células/fisiología
5.
Methods Mol Biol ; 2849: 1-15, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38904915

RESUMEN

Studying human skin biology can aid in comprehending the pathophysiology of skin diseases and developing novel cell-based therapies, including tissue engineering approaches. This chapter provides a comprehensive guide of methods to determine human skin samples from the perspective of their cellular compositions. We describe as useful technique the histological analysis of tissue sections. We further illustrate the biological characterization of isolated and cultured basal and suprabasal interfollicular keratinocytes by cell sorting, cytospin immunostaining, colony forming efficiency, and long-term dermo-epidermal organotypic cultures.


Asunto(s)
Separación Celular , Células Epidérmicas , Queratinocitos , Células Madre , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Separación Celular/métodos , Células Madre/citología , Células Madre/metabolismo , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Piel/citología , Epidermis/metabolismo , Ingeniería de Tejidos/métodos , Diferenciación Celular
6.
Dev Growth Differ ; 66(5): 308-319, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38761018

RESUMEN

One of the major functions of the semaphorin signaling system is the regulation of cell shape. In the nematode Caenorhabditis elegans, membrane-bound semaphorins SMP-1/2 (SMPs) regulate the morphology of epidermal cells via their receptor plexin, PLX-1. In the larval male tail of the SMP-PLX-1 signaling mutants, the border between two epidermal cells, R1.p and R2.p, is displaced anteriorly, resulting in the anterior displacement of the anterior-most ray, ray 1, in the adult male. To elucidate how the intercellular signaling mediated by SMPs regulates the position of the intercellular border, we performed mosaic gene expression analyses by using infrared laser-evoked gene operator (IR-LEGO). We show that PLX-1 expressed in R1.p and SMP-1 expressed in R2.p are required for the proper positioning of ray 1. The result suggests that SMP signaling promotes extension, rather than retraction, of R1.p. This is in contrast to a previous finding that SMPs mediate inhibition of cell extension of vulval precursor cells, another group of epidermal cells of C. elegans, indicating the context dependence of cell shape control via the semaphorin signaling system.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Epidermis , Semaforinas , Animales , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Semaforinas/metabolismo , Semaforinas/genética , Epidermis/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Transducción de Señal , Comunicación Celular , Células Epidérmicas/metabolismo , Células Epidérmicas/citología , Masculino
7.
Methods Mol Biol ; 2849: 17-30, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38700832

RESUMEN

Epidermal stem cells, located in the skin, together with keratinocytes are transplanted in regenerative therapies, e.g., for the treatment of burns or other wounds. Here, we describe the protocol of their enzymatic isolation from human skin. It includes separation of the epidermis form the dermis by incubation with dispase followed by cell isolation for epidermis by digestion with trypsin. Cell isolated with this method can be seeded on collagen IV-coated dishes. The methods of analysis of epidermal stem cells markers (e.g., CD71, CD29) with flow cytometry and RT-PCR are also included.


Asunto(s)
Biomarcadores , Separación Celular , Colágeno Tipo IV , Células Epidérmicas , Citometría de Flujo , Células Madre , Humanos , Citometría de Flujo/métodos , Separación Celular/métodos , Células Epidérmicas/metabolismo , Células Epidérmicas/citología , Células Madre/metabolismo , Células Madre/citología , Colágeno Tipo IV/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Antígenos CD/metabolismo , Antígenos CD/genética , Endopeptidasas
8.
Nat Commun ; 15(1): 3366, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684678

RESUMEN

Autologous skin grafting is a standard treatment for skin defects such as burns. No artificial skin substitutes are functionally equivalent to autologous skin grafts. The cultured epidermis lacks the dermis and does not engraft deep wounds. Although reconstituted skin, which consists of cultured epidermal cells on a synthetic dermal substitute, can engraft deep wounds, it requires the wound bed to be well-vascularized and lacks skin appendages. In this study, we successfully generate complete skin grafts with pluripotent stem cell-derived epidermis with appendages on p63 knockout embryos' dermis. Donor pluripotent stem cell-derived keratinocytes encroach the embryos' dermis by eliminating p63 knockout keratinocytes based on cell-extracellular matrix adhesion mediated cell competition. Although the chimeric skin contains allogenic dermis, it is engraftable as long as autologous grafts. Furthermore, we could generate semi-humanized skin segments by human keratinocytes injection into the amnionic cavity of p63 knockout mice embryos. Niche encroachment opens the possibility of human skin graft production in livestock animals.


Asunto(s)
Dermis , Queratinocitos , Ratones Noqueados , Trasplante de Piel , Animales , Trasplante de Piel/métodos , Queratinocitos/citología , Queratinocitos/trasplante , Humanos , Dermis/citología , Dermis/trasplante , Ratones , Epidermis/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Piel Artificial , Células Epidérmicas/trasplante , Células Epidérmicas/citología , Matriz Extracelular/metabolismo , Piel/citología
9.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38649186

RESUMEN

Numerous long non-coding RNAs (lncRNAs) were shown to have a functional impact on cellular processes such as human epidermal homeostasis. However, the mechanism of action for many lncRNAs remains unclear to date. Here, we report that lncRNA LINC00941 regulates keratinocyte differentiation on an epigenetic level through association with the NuRD complex, one of the major chromatin remodelers in cells. We find that LINC00941 interacts with NuRD-associated MTA2 and CHD4 in human primary keratinocytes. LINC00941 perturbation changes MTA2/NuRD occupancy at bivalent chromatin domains in close proximity to transcriptional regulator genes, including the EGR3 gene coding for a transcription factor regulating epidermal differentiation. Notably, LINC00941 depletion resulted in reduced NuRD occupancy at the EGR3 gene locus, increased EGR3 expression in human primary keratinocytes, and increased abundance of EGR3-regulated epidermal differentiation genes in cells and human organotypic epidermal tissues. Our results therefore indicate a role of LINC00941/NuRD in repressing EGR3 expression in non-differentiated keratinocytes, consequentially preventing premature differentiation of human epidermal tissues.


Asunto(s)
Diferenciación Celular , Epidermis , Histona Desacetilasas , Queratinocitos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , ARN Largo no Codificante , Proteínas Represoras , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Diferenciación Celular/genética , Queratinocitos/metabolismo , Queratinocitos/citología , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Epidermis/metabolismo , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Proteína 3 de la Respuesta de Crecimiento Precoz/genética , Proteína 3 de la Respuesta de Crecimiento Precoz/metabolismo , Epigénesis Genética , Células Epidérmicas/metabolismo , Células Epidérmicas/citología , Cromatina/metabolismo , Cromatina/genética , Regulación de la Expresión Génica , Células Cultivadas
10.
Methods Mol Biol ; 2849: 45-54, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38407797

RESUMEN

The mammalian integumentary system, including skin and its appendages, serves as a protective barrier for the body. During development, skin epidermis undergoes rapid cell division and differentiation to form multiple stratified layers of keratinocytes. Concurrently the epidermis also gives rise to hair follicles that invaginate into the dermis. In adult skin, the hair follicle undergoes cyclic regeneration fueled by hair follicle stem cells located in the bulge. Three-dimensional and high-resolution imaging of these structures using whole-mount immunofluorescent staining allows to better characterize epidermal progenitors and stem cells.


Asunto(s)
Epidermis , Técnica del Anticuerpo Fluorescente , Folículo Piloso , Animales , Folículo Piloso/citología , Folículo Piloso/metabolismo , Ratones , Epidermis/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Animales Recién Nacidos , Coloración y Etiquetado/métodos , Piel/citología , Piel/metabolismo , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo
11.
Methods Mol Biol ; 2849: 173-183, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38376750

RESUMEN

Diabetic foot ulcers (DFUs) pose a significant threat to the health and well-being of individuals with diabetes, often leading to lower limb amputations. Fortunately, epidermal stem cell therapy offers hope for improving the treatment of DFUs. By leveraging 3D culture techniques, the scalability of stem cell manufacturing can be greatly enhanced. In particular, using bioactive materials and scaffolds can promote the healing potential of cells, enhance their proliferation, and facilitate their survival. Furthermore, 3D tissue-mimicking cultures can accurately replicate the complex interactions between cells and extracellular matrix, thereby ensuring that the stem cells are primed for therapeutic application. To ensure the safety and quality of these stem cells, it is essential to adhere to good manufacturing practice (GMP) principles during cultivation. This chapter provides a comprehensive overview of the step-by-step process for GMP-based 3D epidermal stem cell cultivation, thus laying the groundwork for developing reliable regenerative medicine therapies.


Asunto(s)
Pie Diabético , Células Madre , Pie Diabético/terapia , Pie Diabético/patología , Humanos , Células Madre/citología , Células Epidérmicas/citología , Técnicas de Cultivo Tridimensional de Células/métodos , Técnicas de Cultivo de Célula/métodos , Andamios del Tejido/química , Medicina Regenerativa/métodos , Trasplante de Células Madre/métodos , Proliferación Celular , Cicatrización de Heridas , Células Cultivadas , Diferenciación Celular
12.
Cell Tissue Bank ; 24(1): 253-264, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35986799

RESUMEN

To investigate the effect of human adipose tissue-derived multilineage-differentiating stress-enduring (Muse) cells on the oxidative stress injury of human epidermal melanocytes (HEMs) in vitro. HEMs were treated with H2O2 to establish an oxidative stress injury model and then were co-cultured with adipose tissue-derived Muse cells. Immunohistochemistry, flow cytometry and Western blotting were used to assess changes in autophagy flux, apoptosis, expression of melanin synthesis related proteins and proliferation of melanocytes. Our findings demonstrate that co-culture with Muse cells significantly increased the tolerance of HEMs to oxidative stress, enhanced autophagy flux and reduced apoptosis. The expression of proteins related to the formation of melanin increased as did cell proliferation. Treatment with the autophagy inhibitor, 3-methyladenine (3MA), partially counteracted the improvement of oxidative stress tolerance in melanocytes elicited by co-culture with Muse cells. Muse cells promote autophagy and oxidative stress tolerance of melanocytes.


Asunto(s)
Tejido Adiposo , Autofagia , Melanocitos , Células Madre Mesenquimatosas , Tejido Adiposo/citología , Humanos , Femenino , Células Epidérmicas/citología , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanocitos/patología , Estrés Oxidativo , Apoptosis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Técnicas de Cocultivo , Exosomas/metabolismo , Peróxido de Hidrógeno/farmacología , Proliferación Celular , Adulto
13.
Stem Cell Res Ther ; 13(1): 314, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35841007

RESUMEN

BACKGROUND: The biological activity and regenerative medicine of bone marrow mesenchymal stem cells (BMSCs) have been focal topics in the broad fields of diabetic wound repair. However, the molecular mechanisms are still largely elusive for other cellular processes that are regulated during BMSC treatment. Our previous studies have shown that hypoxia is not only a typical pathological phenomenon of wounds but also exerts a vital regulatory effect on cellular bioactivity. In this study, the beneficial effects of hypoxic BMSCs on the cellular behaviors of epidermal cells and diabetic wound healing were investigated. METHOD: The viability and secretion ability of hypoxic BMSCs were detected. The autophagy, proliferation and migration of HaCaT cells cultured with hypoxic BMSCs-derived conditioned medium were assessed by estimating the expression of autophagy-related proteins, MTS, EdU proliferation and scratch assays. And the role of the SMAD signaling pathway during hypoxic BMSC-evoked HaCaT cell autophagy was explored through a series of in vitro gain- and loss-of-function experiments. Finally, the therapeutic effects of hypoxic BMSCs were evaluated using full-thickness cutaneous diabetic wound model. RESULTS: First, we demonstrated that hypoxic conditions intensify HIF-1α-mediated TGF-ß1 secretion by BMSCs. Then, the further data revealed that BMSC-derived TGF-ß1 was responsible for the activation of epidermal cell autophagy, which contributed to the induction of epidermal cell proliferation and migration. Here, the SMAD signaling pathway was identified as downstream of BMSC-derived TGF-ß1 to regulate HaCaT cell autophagy. Moreover, the administration of BMSCs to diabetic wounds increased epidermal autophagy and the rate of re-epithelialization, leading to accelerated healing, and these effects were significantly attenuated, accompanied by the downregulation of Smad2 phosphorylation levels due to TGF-ß1 interference in BMSCs. CONCLUSION: In this report, we present evidence that uncovers a previously unidentified role of hypoxic BMSCs in regulating epidermal cell autophagy. The findings demonstrate that BMSC-based treatment by restoring epidermal cell autophagy could be an attractive therapeutic strategy for diabetic wounds and that the process is mediated by the HIF-1α/TGF-ß1/SMAD pathway.


Asunto(s)
Diabetes Mellitus , Subunidad alfa del Factor 1 Inducible por Hipoxia , Células Madre Mesenquimatosas , Proteínas Smad , Factor de Crecimiento Transformador alfa , Cicatrización de Heridas , Autofagia , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Diabetes Mellitus/terapia , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas/fisiología
14.
Nature ; 606(7912): 188-196, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35585237

RESUMEN

Proper ectodermal patterning during human development requires previously identified transcription factors such as GATA3 and p63, as well as positional signalling from regional mesoderm1-6. However, the mechanism by which ectoderm and mesoderm factors act to stably pattern gene expression and lineage commitment remains unclear. Here we identify the protein Gibbin, encoded by the Xia-Gibbs AT-hook DNA-binding-motif-containing 1 (AHDC1) disease gene7-9, as a key regulator of early epithelial morphogenesis. We find that enhancer- or promoter-bound Gibbin interacts with dozens of sequence-specific zinc-finger transcription factors and methyl-CpG-binding proteins to regulate the expression of mesoderm genes. The loss of Gibbin causes an increase in DNA methylation at GATA3-dependent mesodermal genes, resulting in a loss of signalling between developing dermal and epidermal cell types. Notably, Gibbin-mutant human embryonic stem-cell-derived skin organoids lack dermal maturation, resulting in p63-expressing basal cells that possess defective keratinocyte stratification. In vivo chimeric CRISPR mouse mutants reveal a spectrum of Gibbin-dependent developmental patterning defects affecting craniofacial structure, abdominal wall closure and epidermal stratification that mirror patient phenotypes. Our results indicate that the patterning phenotypes seen in Xia-Gibbs and related syndromes derive from abnormal mesoderm maturation as a result of gene-specific DNA methylation decisions.


Asunto(s)
Proteínas de Unión al ADN , Epitelio , Regulación del Desarrollo de la Expresión Génica , Mesodermo , Morfogénesis , Animales , Humanos , Ratones , Dermis/citología , Dermis/embriología , Dermis/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Ectodermo/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/embriología , Factor de Transcripción GATA3 , Mesodermo/metabolismo , Mutación , Organoides , Transactivadores , Factores de Transcripción/metabolismo
15.
Oncogene ; 41(24): 3341-3354, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35534539

RESUMEN

It is known that Rho GTPases control different aspects of the biology of skin stem cells (SSCs). However, little information is available on the role of their upstream regulators under normal and tumorigenic conditions in this process. To address this issue, we have used here mouse models in which the activity of guanosine nucleotide exchange factors of the Vav subfamily has been manipulated using both gain- and loss-of-function strategies. These experiments indicate that Vav2 and Vav3 regulate the number, functional status, and responsiveness of hair follicle bulge stem cells. This is linked to gene expression programs related to the reinforcement of the identity and the quiescent state of normal SSCs. By contrast, in the case of cancer stem cells, they promote transcriptomal programs associated with the identity, activation state, and cytoskeletal remodeling. These results underscore the role of these Rho exchange factors in the regulation of normal and tumor epidermal stem cells.


Asunto(s)
Proteínas Proto-Oncogénicas c-vav , Piel , Células Madre , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Epidermis/metabolismo , Folículo Piloso/citología , Folículo Piloso/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Piel/citología , Piel/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Células Madre/citología , Células Madre/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
16.
Sci Adv ; 8(17): eabl8698, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35476447

RESUMEN

Uniquely among mammalian organs, skin is capable of marked size change in adults, yet the mechanisms underlying this notable capacity are unclear. Here, we use a system of controlled tissue expansion in mice to uncover cellular and molecular determinants of skin growth. Through machine learning-guided three-dimensional tissue reconstruction, we capture morphometric changes in growing skin. We find that most growth is driven by the proliferation of the epidermis in response to mechanical tension, with more limited changes in dermal and subdermal compartments. Epidermal growth is achieved through preferential activation and differentiation of Lgr6+ stem cells of the epidermis, driven in part by the Hippo pathway. By single-cell RNA sequencing, we uncover further changes in mechanosensitive and metabolic pathways underlying growth control in the skin. These studies point to therapeutic strategies to enhance skin growth and establish a platform for understanding organ size dynamics in adult mammals.


Asunto(s)
Células Epidérmicas , Receptores Acoplados a Proteínas G , Piel , Células Madre , Animales , Células Epidérmicas/citología , Células Epidérmicas/metabolismo , Epidermis/crecimiento & desarrollo , Epidermis/metabolismo , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Piel/crecimiento & desarrollo , Piel/metabolismo , Células Madre/citología , Células Madre/metabolismo
17.
Sci Rep ; 12(1): 3184, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210511

RESUMEN

Cryopreserved allogeneic cultured epidermis (CE) is used for treating second-degree burn wounds and diabetic foot ulcers; however, the need for cryopreservation limits its use. We have previously reported that CE accelerates wound healing irrespective of its viability and hypothesized that dehydrated CEs lacking living cells may act as an effective wound dressing. We prepared dried CE and investigated its morphological and physical properties and wound-healing effects and compared them with those of cryopreserved CE. Hematoxylin-eosin staining, immunostaining for basement membrane, and electron microscopy revealed that the morphologies of dried CE and cryopreserved CE were comparable and that the membrane structure was not damaged. The breaking strength, modulus of elasticity, and water permeability of dried CE were comparable with those of the cryopreserved CE. Furthermore, the levels of various active cytokines and chemokines in dried CE were comparable with those in cryopreserved CE. Dried CE applied to skin defect in diabetic mice significantly reduced the wound area and increased the new epithelium length 4 and 7 days after implantation, similar to that observed for cryopreserved CE. Consequently, dried CE had similar morphological and physical properties and wound-healing effects compared with those of cryopreserved CE and can be a physiological and versatile wound-dressing.


Asunto(s)
Células Epidérmicas/trasplante , Epidermis/trasplante , Queratinocitos/trasplante , Piel/patología , Cicatrización de Heridas , Animales , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Criopreservación , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Células Epidérmicas/citología , Liofilización , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Piel/metabolismo
19.
Nat Commun ; 13(1): 876, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35169143

RESUMEN

The membrane receptor kinases HAESA and HSL2 recognize a family of IDA/IDL signaling peptides to control cell separation processes in different plant organs. The homologous HSL1 has been reported to regulate epidermal cell patterning by interacting with a different class of signaling peptides from the CLE family. Here we demonstrate that HSL1 binds IDA/IDL peptides with high, and CLE peptides with lower affinity, respectively. Ligand sensing capability and receptor activation of HSL1 require a SERK co-receptor kinase. Crystal structures with IDA/IDLs or with CLE9 reveal that HSL1-SERK1 complex recognizes the entire IDA/IDL signaling peptide, while only parts of CLE9 are bound to the receptor. In contrast, the receptor kinase BAM1 interacts with the entire CLE9 peptide with high affinity and specificity. Furthermore, the receptor tandem BAM1/BAM2 regulates epidermal cell division homeostasis. Consequently, HSL1-IDLs and BAM1/BAM2-CLEs independently regulate cell patterning in the leaf epidermal tissue.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Células Epidérmicas/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Hojas de la Planta/embriología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Arabidopsis , Proteínas de Arabidopsis/genética , Línea Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/genética , Células Sf9 , Nicotiana
20.
Nat Cell Biol ; 24(1): 10-23, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34969962

RESUMEN

Loss of alveolar type 2 cells (AEC2s) and the ectopic appearance of basal cells in the alveoli characterize severe lung injuries such as idiopathic pulmonary fibrosis (IPF). Here we demonstrate that human alveolar type 2 cells (hAEC2s), unlike murine AEC2s, transdifferentiate into basal cells in response to fibrotic signalling in the lung mesenchyme, in vitro and in vivo. Single-cell analysis of normal hAEC2s and mesenchymal cells in organoid co-cultures revealed the emergence of pathologic fibroblasts and basaloid cells previously described in IPF. Transforming growth factor-ß1 and anti-bone morphogenic protein signalling in the organoids promoted transdifferentiation. Trajectory and histologic analyses of both hAEC2-derived organoids and IPF epithelium indicated that hAEC2s transdifferentiate into basal cells through alveolar-basal intermediates that accumulate in proximity to pathologic CTHRC1hi/TGFB1hi fibroblasts. Our study indicates that hAEC2 loss and expansion of alveolar metaplastic basal cells in severe human lung injuries are causally connected through an hAEC2-basal cell lineage trajectory driven by aberrant mesenchyme.


Asunto(s)
Transdiferenciación Celular/fisiología , Células Epiteliales/citología , Fibrosis Pulmonar Idiopática/patología , Queratina-5/metabolismo , Alveolos Pulmonares/citología , Mucosa Respiratoria/citología , Células Epiteliales Alveolares/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Células Cultivadas , Células Epidérmicas/citología , Fibroblastos/citología , Humanos , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Transducción de Señal/fisiología , Análisis de la Célula Individual , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...