Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Nature ; 606(7912): 188-196, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35585237

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA , Epitélio , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma , Morfogênese , Animais , Humanos , Camundongos , Derme/citologia , Derme/embriologia , Derme/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Ectoderma/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Epidérmicas/citologia , Células Epidérmicas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/embriologia , Fator de Transcrição GATA3 , Mesoderma/metabolismo , Mutação , Organoides , Transativadores , Fatores de Transcrição/metabolismo
2.
Dev Biol ; 478: 25-40, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34166654

RESUMO

Skin development and patterning is dependent on factors that regulate the stepwise differentiation of dermal fibroblasts concomitant with dermal-epidermal reciprocal signaling, two processes that are poorly understood. Here we show that dermal EZH2, the methyltransferase enzyme of the epigenetic Polycomb Repressive Complex 2 (PRC2), is a new coordinator of both these processes. Dermal EZH2 activity is present during dermal fibroblast differentiation and is required for spatially restricting Wnt/ß-catenin signaling to reinforce dermal fibroblast cell fate. Later in development, dermal EZH2 regulates the expression of reticular dermal markers and initiation of secondary hair follicles. Embryos lacking dermal Ezh2 have elevated epidermal proliferation and differentiation that can be rescued by small molecule inhibition of retinoic acid (RA) signaling. Together, our study reveals that dermal EZH2 is acting like a rheostat to control the levels of Wnt/ß-catenin and RA signaling to impact fibroblast differentiation cell autonomously and epidermal keratinocyte development non-cell autonomously, respectively.


Assuntos
Derme/citologia , Derme/embriologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epiderme/embriologia , Fibroblastos/citologia , Queratinócitos/citologia , Complexo Repressor Polycomb 2/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Derme/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epiderme/metabolismo , Fibroblastos/metabolismo , Hiperplasia , Queratinócitos/metabolismo , Camundongos , Organogênese , Retinoides/farmacologia , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Tretinoína/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
3.
EMBO J ; 37(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30297530

RESUMO

Endothelial cells line blood and lymphatic vessels and form intercellular junctions, which preserve vessel structure and integrity. The vascular endothelial cadherin, VE-cadherin, mediates endothelial adhesion and is indispensible for blood vessel development and permeability regulation. However, its requirement for lymphatic vessels has not been addressed. During development, VE-cadherin deletion in lymphatic endothelial cells resulted in abortive lymphangiogenesis, edema, and prenatal death. Unexpectedly, inducible postnatal or adult deletion elicited vessel bed-specific responses. Mature dermal lymph vessels resisted VE-cadherin loss and maintained button junctions, which was associated with an upregulation of junctional molecules. Very different, mesenteric lymphatic collectors deteriorated and formed a strongly hyperplastic layer of lymphatic endothelial cells on the mesothelium. This massive hyperproliferation may have been favored by high mesenteric VEGF-C expression and was associated with VEGFR-3 phosphorylation and upregulation of the transcriptional activator TAZ Finally, intestinal lacteals fragmented into cysts or became highly distended possibly as a consequence of the mesenteric defects. Taken together, we demonstrate here the importance of VE-cadherin for lymphatic vessel development and maintenance, which is however remarkably vessel bed-specific.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Derme/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Linfangiogênese , Vasos Linfáticos/metabolismo , Mesentério/embriologia , Animais , Antígenos CD/genética , Caderinas/genética , Células Endoteliais/metabolismo , Deleção de Genes , Camundongos , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Dev Biol ; 455(1): 60-72, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31279726

RESUMO

Salamanders are capable of full-thickness skin regeneration where removal of epidermis, dermis and hypodermis results in scar-free repair. What remains unclear is whether regeneration of these tissues recapitulates the cellular events of skin development or occurs through a process unique to regenerative healing. Unfortunately, information on the post-embryonic development of salamander skin is severely lacking, having focused on compartments or cell types, but never on the skin as a complete organ. By examining coordinated development of the epidermis and dermis in axolotls we establish six distinct stages of skin development (I-VI): I-V for normally paedomorphic adults and a sixth stage following metamorphosis. Raising animals either in isolation (zero density pressure) or in groups (density pressure) we find that skin development progresses as a function of animal size and that density directly effects developmental rate. Using keratins, p63, and proliferative markers, we show that when the dermis transforms into the stratum spongiosum and stratum compactum, keratinocytes differentiate into at least three distinct phenotypes that reveal a cryptic stratification program uncoupled from metamorphosis. Lastly, comparing skin regeneration to skin development, we find that dermal regeneration occurs through a unique process, relying heavily on remodeling of the wound extracellular matrix, rather than proceeding through direct development of a dermal lamella produced by the epidermis. By preventing fibroblast influx into the wound bed using beryllium nitrate, we show that in the absence of fibroblast generated ECM production skin regeneration occurs through an alternate route that recapitulates development.


Assuntos
Desenvolvimento Embrionário/fisiologia , Fibroblastos/fisiologia , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Pele/fisiopatologia , Cicatrização/fisiologia , Ambystoma mexicanum/embriologia , Ambystoma mexicanum/fisiologia , Animais , Derme/embriologia , Derme/metabolismo , Derme/fisiologia , Epiderme/embriologia , Epiderme/metabolismo , Epiderme/fisiologia , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiologia , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Queratinas/metabolismo , Masculino , Pele/embriologia , Pele/lesões , Fatores de Tempo
5.
Development ; 144(10): 1887-1895, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28512199

RESUMO

SOX family proteins SOX2 and SOX18 have been reported as being essential in determining hair follicle type; however, the role they play during development remains unclear. Here, we demonstrate that Sox18 regulates the normal differentiation of the dermal papilla of all hair types. In guard (primary) hair dermal condensate (DC) cells, we identified transient Sox18 in addition to SOX2 expression at E14.5, which allowed fate tracing of primary DC cells until birth. Similarly, expression of Sox18 was detected in the DC cells of secondary hairs at E16.5 and in tertiary hair at E18.5. Dominant-negative Sox18 mutation (opposum) did not prevent DC formation in any hair type. However, it affected dermal papilla differentiation, restricting hair formation especially in secondary and tertiary hairs. This Sox18 mutation also prevented neonatal dermal cells or dermal papilla spheres from inducing hair in regeneration assays. Microarray expression studies identified WNT5A and TNC as potential downstream effectors of SOX18 that are important for epidermal WNT signalling. In conclusion, SOX18 acts as a mesenchymal molecular switch necessary for the formation and function of the dermal papilla in all hair types.


Assuntos
Diferenciação Celular/genética , Folículo Piloso/embriologia , Cabelo/embriologia , Fatores de Transcrição SOXF/fisiologia , Animais , Derme/embriologia , Derme/metabolismo , Embrião de Mamíferos , Células Epidérmicas , Epiderme/embriologia , Feminino , Genes Dominantes , Genes de Troca/fisiologia , Cabelo/metabolismo , Folículo Piloso/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição SOXF/genética
6.
Development ; 143(22): 4193-4202, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27697906

RESUMO

Cellular migrations through constricted spaces are a crucial aspect of many developmental and disease processes including hematopoiesis, inflammation and metastasis. A limiting factor in these events is nuclear deformation. Here, we establish an in vivo model in which nuclei can be visualized while moving through constrictions and use it to elucidate mechanisms for nuclear migration. C. elegans hypodermal P-cell larval nuclei traverse a narrow space that is about 5% their width. This constriction is blocked by fibrous organelles, structures that pass through P cells to connect the muscles to cuticle. Fibrous organelles are removed just prior to nuclear migration, when nuclei and lamins undergo extreme morphological changes to squeeze through the space. Both actin and microtubule networks are organized to mediate nuclear migration. The LINC complex, consisting of the SUN protein UNC-84 and the KASH protein UNC-83, recruits dynein and kinesin-1 to the nuclear surface. Both motors function in P-cell nuclear migration, but dynein, functioning through UNC-83, plays a more central role as nuclei migrate towards minus ends of polarized microtubule networks. Thus, the nucleoskeleton and cytoskeleton are coordinated to move nuclei through constricted spaces.


Assuntos
Citoesqueleto de Actina/metabolismo , Caenorhabditis elegans , Núcleo Celular/metabolismo , Derme/embriologia , Derme/metabolismo , Microtúbulos/metabolismo , Actinas/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Biológico , Padronização Corporal , Caenorhabditis elegans/citologia , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Citoesqueleto/metabolismo , Derme/ultraestrutura , Embrião não Mamífero
7.
Nature ; 504(7479): 277-281, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24336287

RESUMO

Fibroblasts are the major mesenchymal cell type in connective tissue and deposit the collagen and elastic fibres of the extracellular matrix (ECM). Even within a single tissue, fibroblasts exhibit considerable functional diversity, but it is not known whether this reflects the existence of a differentiation hierarchy or is a response to different environmental factors. Here we show, using transplantation assays and lineage tracing in mice, that the fibroblasts of skin connective tissue arise from two distinct lineages. One forms the upper dermis, including the dermal papilla that regulates hair growth and the arrector pili muscle, which controls piloerection. The other forms the lower dermis, including the reticular fibroblasts that synthesize the bulk of the fibrillar ECM, and the preadipocytes and adipocytes of the hypodermis. The upper lineage is required for hair follicle formation. In wounded adult skin, the initial wave of dermal repair is mediated by the lower lineage and upper dermal fibroblasts are recruited only during re-epithelialization. Epidermal ß-catenin activation stimulates the expansion of the upper dermal lineage, rendering wounds permissive for hair follicle formation. Our findings explain why wounding is linked to formation of ECM-rich scar tissue that lacks hair follicles. They also form a platform for discovering fibroblast lineages in other tissues and for examining fibroblast changes in ageing and disease.


Assuntos
Linhagem da Célula , Fibroblastos/citologia , Pele/citologia , Pele/crescimento & desenvolvimento , Cicatrização/fisiologia , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Derme/anatomia & histologia , Derme/citologia , Derme/embriologia , Derme/crescimento & desenvolvimento , Feminino , Fibroblastos/transplante , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Músculo Liso/citologia , Músculo Liso/metabolismo , Pele/anatomia & histologia , Pele/embriologia , beta Catenina/metabolismo
8.
Int J Mol Sci ; 20(16)2019 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-31412620

RESUMO

Two mesenchymal zinc transporters, ZIP7 and ZIP13, play critical roles in dermal development. ZIP7 and ZIP13 are the closest among the conserved mammalian zinc transporters. However, whether their functions are complementary remains a controversial issue. In the present study, we found that the expression of ZIP13, but not ZIP7, is elevated by transforming growth factor beta (TGF-ß) treatment, indicating that TGF-ß-mediated ZIP13 amplification is crucial for collagen production during dermal development. Genome-wide gene expression analysis revealed that ~26% of genes are dependent on either ZIP7 or ZIP13, which is greater than the ~17% of genes dependent on both of them. ZIP7 depletion induces endoplasmic reticulum (ER) stress in mesenchymal stem cells, resulting in significant inhibition of fibrogenic differentiation. However, ZIP13 depletion does not induce ER stress. Though both ZIP7 and ZIP13 contain traditional ER signal peptides for their intracellular localization, their distributions are distinct. When ZIP7 and ZIP13 are coexpressed, their localizations are distinct; ZIP7 is located on the ER, but ZIP13 is located on both the ER and Golgi, indicating that only ZIP13 is a zinc gatekeeper on the Golgi. Our data illustrate that the different actions of ZIP7 and ZIP13 are crucial for dermal development.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Derme/embriologia , Derme/metabolismo , Organogênese/genética , Zinco/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Genoma , Estudo de Associação Genômica Ampla , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos
9.
Adv Gerontol ; 32(3): 357-363, 2019.
Artigo em Russo | MEDLINE | ID: mdl-31512421

RESUMO

The aim of this work was to examine the content of Piezo1 in fibroblasts and blood vessels of human dermis from the development until deep aging (from 20 weeks of pregnancy until 85 years old), and defining of a role of Piezo1 in age-dependent changes in the number of fibroblasts and blood vessels in the dermis. Piezo1, proliferating cells nuclear antigen (PCNA), endothelial cells marker CD31 were detected with indirect immunohistochemical technique. Results showed that a portion of fibroblasts with positive staining for Piezo1 in the dermis is decreased from 20 weeks of pregnancy to 40 years old. Percent of Piezo1 positive fibroblasts in dermis is increased sufficiently since 41 years old until 60-85 years old group. The content of Piezo1 in blood vessels in the human dermis is decreased sufficiently from 20 weeks of pregnancy until 40 years old. Age-related changes in the content of Piezo1 in fibroblasts and blood vessels is not associated with an age-related decrease in total number and percent of PCNA positive fibroblasts, the number of blood vessels in the dermis.


Assuntos
Vasos Sanguíneos , Derme , Fibroblastos , Canais Iônicos , Envelhecimento da Pele , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Vasos Sanguíneos/fisiologia , Criança , Pré-Escolar , Derme/irrigação sanguínea , Derme/citologia , Derme/embriologia , Derme/crescimento & desenvolvimento , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Lactente , Canais Iônicos/metabolismo , Masculino , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Envelhecimento da Pele/fisiologia
10.
Dev Biol ; 413(1): 60-9, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26968353

RESUMO

Sensory dendrite morphogenesis is directed by intrinsic and extrinsic factors. The extracellular environment plays instructive roles in patterning dendrite growth and branching. However, the molecular mechanism is not well understood. In Caenorhabditis elegans, the proprioceptive neuron PVD forms highly branched sensory dendrites adjacent to the hypodermis. We report that receptor tyrosine phosphatase CLR-1 functions in the hypodermis to pattern the PVD dendritic branches. Mutations in clr-1 lead to loss of quaternary branches, reduced secondary branches and increased ectopic branches. CLR-1 is necessary for the dendrite extension but not for the initial filopodia formation. Its role is dependent on the intracellular phosphatase domain but not the extracellular adhesion domain, indicating that it functions through dephosphorylating downstream factors but not through direct adhesion with neurons. Genetic analysis reveals that clr-1 also functions in parallel with SAX-7/DMA-1 pathway to control PVD primary dendrite development. We provide evidence of a new environmental factor for PVD dendrite morphogenesis.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Dendritos/metabolismo , Proteínas Tirosina Fosfatases Semelhantes a Receptores/fisiologia , Pele/embriologia , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Derme/embriologia , Proteínas de Fluorescência Verde/metabolismo , Mutação , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Pseudópodes/metabolismo , Células Receptoras Sensoriais/metabolismo , Transgenes
11.
Genesis ; 54(8): 415-30, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27265328

RESUMO

Embryonic dermal fibroblasts in the skin have the exceptional ability to initiate hair follicle morphogenesis and contribute to scarless wound healing. Activation of the Wnt signaling pathway is critical for dermal fibroblast fate selection and hair follicle induction. In humans, mutations in Wnt pathway components and target genes lead to congenital focal dermal hypoplasias with diminished hair. The gene expression signature of embryonic dermal fibroblasts during differentiation and its dependence on Wnt signaling is unknown. Here we applied Shannon entropy analysis to identify the gene expression signature of mouse embryonic dermal fibroblasts. We used available human DNase-seq and histone modification ChiP-seq data on various cell-types to demonstrate that genes in the fibroblast cell identity signature can be epigenetically repressed in other cell-types. We found a subset of the signature genes whose expression is dependent on Wnt/ß-catenin activity in vivo. With our approach, we have defined and validated a statistically derived gene expression signature that may mediate dermal fibroblast identity and function in development and disease. genesis 54:415-430, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular , Derme/citologia , Fibroblastos/metabolismo , Transcriptoma , Via de Sinalização Wnt/genética , Animais , Derme/embriologia , Fibroblastos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos
12.
Proc Natl Acad Sci U S A ; 110(29): 11899-904, 2013 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-23818600

RESUMO

Classical hypotheses regarding the evolutionary origin of paired appendages propose transformation of precursor structures (gill arches and lateral fin folds) into paired fins. During development, gnathostome paired appendages form as outgrowths of body wall somatopleure, a tissue composed of somatic lateral plate mesoderm (LPM) and overlying ectoderm. In amniotes, LPM contributes connective tissue to abaxial musculature and forms ventrolateral dermis of the interlimb body wall. The phylogenetic distribution of this character is uncertain because lineage analyses of LPM have not been generated in anamniotes. We focus on the evolutionary history of the somatopleure to gain insight into the tissue context in which paired fins first appeared. Lampreys diverged from other vertebrates before the acquisition of paired fins and provide a model for investigating the preappendicular condition. We present vital dye fate maps that suggest the somatopleure is eliminated in lamprey as the LPM is separated from the ectoderm and sequestered to the coelomic linings during myotome extension. We also examine the distribution of postcranial mesoderm in catshark and axolotl. In contrast to lamprey, our findings support an LPM contribution to the trunk body wall of these taxa, which is similar to published data for amniotes. Collectively, these data lead us to hypothesize that a persistent somatopleure in the lateral body wall is a gnathostome synapomorphy, and the redistribution of LPM was a key step in generating the novel developmental module that ultimately produced paired fins. These embryological criteria can refocus arguments on paired fin origins and generate hypotheses testable by comparative studies on the source, sequence, and extent of genetic redeployment.


Assuntos
Nadadeiras de Animais/embriologia , Evolução Biológica , Derme/embriologia , Lampreias/embriologia , Mesoderma/embriologia , Ambystoma mexicanum/embriologia , Animais , Linhagem da Célula/fisiologia , Crioultramicrotomia , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Filogenia , Tubarões/embriologia
13.
Exp Dermatol ; 24(6): 468-70, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25708924

RESUMO

Embryonic hair follicle (HF) induction and formation is dependent on signalling crosstalk between the dermis and specialized dermal condensates on the mesenchymal side and epidermal cells and incipient placodes on the epithelial side, but the precise nature and succession of signals remain unclear. Platelet-derived growth factor (PDGF) signalling is involved in the development of several organs and the maintenance of adult tissues, including HF regeneration in the hair cycle. As both PDGF receptors, PDGFRα and PDGFRß, are expressed in embryonic dermis and dermal condensates, we explored in this study the role of PDGF signalling in HF induction and formation in the developing skin mesenchyme. We conditionally ablated both PDGF receptors with Tbx18(Cre) in early dermal condensates before follicle formation, and with Prx1-Cre broadly in the ventral dermis prior to HF induction. In both PDGFR double mutants, HF induction and formation ensued normally, and the pattern of HF formation and HF numbers were unaffected. These data demonstrate that mesenchymal PDGF signalling, either in the specialized niche or broadly in the dermis, is dispensable for HF induction and formation.


Assuntos
Derme/embriologia , Folículo Piloso/embriologia , Morfogênese/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/fisiologia , Animais , Derme/citologia , Derme/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/fisiologia , Camundongos , Camundongos Mutantes , Modelos Animais , Morfogênese/genética , Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/genética
14.
Development ; 138(18): 3943-54, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21862558

RESUMO

We aim to evaluate environmental and genetic effects on the expansion/proliferation of committed single cells during embryonic development, using melanoblasts as a paradigm to model this phenomenon. Melanoblasts are a specific type of cell that display extensive cellular proliferation during development. However, the events controlling melanoblast expansion are still poorly understood due to insufficient knowledge concerning their number and distribution in the various skin compartments. We show that melanoblast expansion is tightly controlled both spatially and temporally, with little variation between embryos. We established a mathematical model reflecting the main cellular mechanisms involved in melanoblast expansion, including proliferation and migration from the dermis to epidermis. In association with biological information, the model allows the calculation of doubling times for melanoblasts, revealing that dermal and epidermal melanoblasts have short but different doubling times. Moreover, the number of trunk founder melanoblasts at E8.5 was estimated to be 16, a population impossible to count by classical biological approaches. We also assessed the importance of the genetic background by studying gain- and loss-of-function ß-catenin mutants in the melanocyte lineage. We found that any alteration of ß-catenin activity, whether positive or negative, reduced both dermal and epidermal melanoblast proliferation. Finally, we determined that the pool of dermal melanoblasts remains constant in wild-type and mutant embryos during development, implying that specific control mechanisms associated with cell division ensure half of the cells at each cell division to migrate from the dermis to the epidermis. Modeling melanoblast expansion revealed novel links between cell division, cell localization within the embryo and appropriate feedback control through ß-catenin.


Assuntos
Diferenciação Celular , Crescimento e Desenvolvimento/fisiologia , Melanócitos/fisiologia , Modelos Biológicos , Modelos Teóricos , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , Derme/citologia , Derme/embriologia , Embrião de Mamíferos , Células Epidérmicas , Epiderme/embriologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
15.
Development ; 138(19): 4155-66, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21852400

RESUMO

The plane of cell divisions is pivotal for differential fate acquisition. Dermomyotome development provides an excellent system with which to investigate the link between these processes. In the central sheet of the early dermomyotome, single epithelial cells divide with a planar orientation. Here, we report that in the avian embryo, in addition to self-renewing, a subset of progenitors translocates into the myotome where they generate differentiated myocytes. By contrast, in the late epithelium, individual progenitors divide perpendicularly to produce both mitotic myoblasts and dermis. To examine whether spindle orientations influence fate segregation, early planar divisions were randomized and/or shifted to a perpendicular orientation by interfering with LGN function or by overexpressing inscuteable. Clones derived from single transfected cells exhibited an enhanced proportion of mixed dermomyotome/myotome progeny at the expense of `like' daughter cells in either domain. Loss of LGN or Gαi1 function in the late epithelium randomized otherwise perpendicular mitoses and favored muscle development at the expense of dermis. Hence, LGN-dependent early planar divisions are required for the proper allocation of progenitors into either dermomyotome or myotome, whereas late perpendicular divisions are necessary for the normal balance between muscle and dermis production.


Assuntos
Derme/citologia , Derme/embriologia , Células Musculares/citologia , Músculos/citologia , Músculos/embriologia , Somitos/citologia , Animais , Asparagina/química , Diferenciação Celular/fisiologia , Divisão Celular , Linhagem da Célula , Coturnix/embriologia , Derme/metabolismo , Eletroporação , Epitélio/patologia , Regulação da Expressão Gênica no Desenvolvimento , Glicina/química , Leucina/química , Mitose , Morfogênese , Músculos/metabolismo , Transfecção
16.
Development ; 137(23): 3973-84, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20980404

RESUMO

Cranial dermis develops from cephalic mesoderm and neural crest cells, but what signal(s) specifies the dermal lineage is unclear. Using genetic tools to fate map and manipulate a cranial mesenchymal progenitor population in the supraorbital region, we show that the dermal progenitor cells beneath the surface ectoderm process canonical Wnt signaling at the time of specification. We show that Wnt signaling/ß-catenin is absolutely required and sufficient for Dermo1 expression and dermal cell identity in the cranium. The absence of the Wnt signaling cue leads to formation of cartilage in craniofacial and ventral trunk regions at the expense of dermal and bone lineages. Dermo1 can be a direct transcription target and may mediate the functional role of Wnt signaling in dermal precursors. This study reveals a lineage-specific role of canonical Wnt signaling/ß-catenin in promoting dermal cell fate in distinct precursor populations.


Assuntos
Derme/citologia , Proteínas Repressoras/metabolismo , Transdução de Sinais , Crânio/embriologia , Proteína 1 Relacionada a Twist/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Biomarcadores/metabolismo , Padronização Corporal , Cartilagem/citologia , Cartilagem/metabolismo , Diferenciação Celular , Linhagem da Célula , Derme/embriologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Elementos Facilitadores Genéticos/genética , Proteínas de Homeodomínio/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Proteínas Repressoras/genética , Fatores de Transcrição SOX9/metabolismo , Crânio/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fator de Transcrição 4 , Proteína 1 Relacionada a Twist/genética
17.
Development ; 136(16): 2815-23, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19605494

RESUMO

The dermal papilla comprises the specialised mesenchymal cells at the base of the hair follicle. Communication between dermal papilla cells and the overlying epithelium is essential for differentiation of the hair follicle lineages. We report that Sox2 is expressed in all dermal papillae at E16.5, but from E18.5 onwards expression is confined to a subset of dermal papillae. In postnatal skin, Sox2 is only expressed in the dermal papillae of guard/awl/auchene follicles, whereas CD133 is expressed both in guard/awl/auchene and in zigzag dermal papillae. Using transgenic mice that express GFP under the control of the Sox2 promoter, we isolated Sox2(+) (GFP(+)) CD133(+) cells and compared them with Sox2(-) (GFP(-)) CD133(+) dermal papilla cells. In addition to the 'core' dermal papilla gene signature, each subpopulation expressed distinct sets of genes. GFP(+) CD133(+) cells had upregulated Wnt, FGF and BMP pathways and expressed neural crest markers. In GFP(-) CD133(+) cells, the hedgehog, IGF, Notch and integrin pathways were prominent. In skin reconstitution assays, hair follicles failed to form when dermis was depleted of both GFP(+) CD133(+) and GFP(-) CD133(+) cells. In the absence of GFP(+) CD133(+) cells, awl/auchene hairs failed to form and only zigzag hairs were found. We have thus demonstrated a previously unrecognised heterogeneity in dermal papilla cells and shown that Sox2-positive cells specify particular hair follicle types.


Assuntos
Derme , Epiderme , Folículo Piloso , Fatores de Transcrição SOXB1/metabolismo , Antígeno AC133 , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Derme/citologia , Derme/embriologia , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Células Epidérmicas , Epiderme/embriologia , Epiderme/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Glicoproteínas/genética , Glicoproteínas/metabolismo , Folículo Piloso/citologia , Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/genética , Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição SOXB1/genética , Transdução de Sinais/fisiologia
18.
J Anat ; 221(2): 115-20, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22606994

RESUMO

Somites compartmentalize into a dorsal epithelial dermomyotome and a ventral mesenchymal sclerotome. While sclerotomes give rise to vertebrae and intervertebral discs, dermomyotomes contribute to skeletal muscle and epaxial dermis. Bone morphogenetic protein (BMP)-signals from the lateral mesoderm induce the lateral portion of the dermomyotome to form chondrogenic precursor cells, forming the cartilage of the scapula blade. The fact that BMPs are expressed in the roof plate of the neural tube where they induce cartilage formation led to the question why cells migrating from the medial part of the dermomyotome do not undergo chondrogenic differentiation and do not contribute to the dorsal part of the vertebrae. In the present study, we traced dermomyotomal derivatives by using the quail-chick marker technique. Our study reveals a temporal sequence in the formation of the vertebral cartilage and the midline dermis. The dorsal mesenchyme overlying the roof plate of the neural tube is formed prior to the de-epithelialization of the dermomyotome. Dermomyotomal cells start to migrate medially into the sub-ectodermal space to form the midline dermis after chondrogenesis of the dorsal mesenchyme has occurred. This time delay between chondrogenesis of the dorsal vertebra and dermal formation allows an undisturbed development of these two tissue components within a narrow region of the embryo.


Assuntos
Embrião de Galinha/crescimento & desenvolvimento , Derme/embriologia , Coluna Vertebral/embriologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Cartilagem/embriologia , Codorniz/embriologia , Fatores de Tempo
19.
J Cell Biol ; 177(3): 501-13, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17485490

RESUMO

Integrin-linked kinase (ILK) links integrins to the actin cytoskeleton and is believed to phosphorylate several target proteins. We report that a keratinocyte-restricted deletion of the ILK gene leads to epidermal defects and hair loss. ILK-deficient epidermal keratinocytes exhibited a pronounced integrin-mediated adhesion defect leading to epidermal detachment and blister formation, disruption of the epidermal-dermal basement membrane, and the translocation of proliferating, integrin-expressing keratinocytes to suprabasal epidermal cell layers. The mutant hair follicles were capable of producing hair shaft and inner root sheath cells and contained stem cells and generated proliferating progenitor cells, which were impaired in their downward migration and hence accumulated in the outer root sheath and failed to replenish the hair matrix. In vitro studies with primary ILK-deficient keratinocytes attributed the migration defect to a reduced migration velocity and an impaired stabilization of the leading-edge lamellipodia, which compromised directional and persistent migration. We conclude that ILK plays important roles for epidermis and hair follicle morphogenesis by modulating integrin-mediated adhesion, actin reorganization, and plasma membrane dynamics in keratinocytes.


Assuntos
Movimento Celular , Folículo Piloso/embriologia , Queratinócitos/metabolismo , Organogênese , Proteínas Serina-Treonina Quinases/metabolismo , Células-Tronco/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Membrana Basal/citologia , Membrana Basal/embriologia , Vesícula/genética , Vesícula/metabolismo , Adesão Celular/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Movimento Celular/genética , Proliferação de Células , Derme/citologia , Derme/embriologia , Feminino , Folículo Piloso/citologia , Queratinócitos/citologia , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Organogênese/genética , Proteínas Serina-Treonina Quinases/deficiência , Pseudópodes/genética , Pseudópodes/metabolismo , Células-Tronco/citologia
20.
Adv Gerontol ; 25(4): 604-11, 2012.
Artigo em Russo | MEDLINE | ID: mdl-23734504

RESUMO

There are morphofunctional zones in organism tissues, where proliferation and differentiation processes occur. Daughter cells are differentiated in the electric field excited by 12 mother and daughter cell pairs, turned out at cambial cell division. With aging, the cambial cell number is reduced to 7, close to thresholds level (6 cells), at which the differentiation of daughter cells is absent. The depression of cambial cell number with aging is connected with the work of another morphofunctional zone--the hypothalamus, which is the major center of vegetative regulation and initially has very high RhoA activity, which has been established in embryogenesis. Estrogens, influencing over the hypothalamus and activating Src kinase in its nuclei, reduce the level of RhoA activity, including SCN, responsible for many biorhythms of an organism. As a result, the hyperestrogenemia and therefore a connective tissue at first occur. Then there happens a hypoestrogenemia that leads to sharp falling of proliferative activity of cells, causing the depression of cambial cell number and possibility of a malignant tumor development. Along with this, there are the deep lesions of hormone regulation, leading to some lethal diseases. Thus, the RhoA increasing in hypothalamus and especially in SCN circadian rhythm can counteract the Src kinase intensifying and prevent the processes connected with this.


Assuntos
Envelhecimento , Derme/embriologia , Derme/crescimento & desenvolvimento , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Morfogênese , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Diferenciação Celular , Proliferação de Células , Derme/citologia , Derme/metabolismo , Estrogênios/metabolismo , Humanos , Hipotálamo/citologia , Hipotálamo/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA