Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Invest Dermatol ; 142(6): 1597-1606.e9, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34808238

RESUMO

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.


Assuntos
Dipeptidil Peptidase 4 , Dermatopatias , Animais , Dipeptidil Peptidase 4/genética , Fibroblastos/metabolismo , Fibrose , Camundongos , Pele/patologia , Dermatopatias/genética , Dermatopatias/patologia , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo
2.
Nat Commun ; 10(1): 5023, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31685822

RESUMO

Melanoma, the deadliest skin cancer, remains largely incurable at advanced stages. Currently, there is a lack of animal models that resemble human melanoma initiation and progression. Recent studies using a Tyr-CreER driven mouse model have drawn contradictory conclusions about the potential of melanocyte stem cells (McSCs) to form melanoma. Here, we employ a c-Kit-CreER-driven model that specifically targets McSCs to show that oncogenic McSCs are a bona fide source of melanoma that expand in the niche, and then establish epidermal melanomas that invade into the underlying dermis. Further, normal Wnt and Endothelin niche signals during hair anagen onset are hijacked to promote McSC malignant transformation during melanoma induction. Finally, molecular profiling reveals strong resemblance of murine McSC-derived melanoma to human melanoma in heterogeneity and gene signatures. These findings provide experimental validation of the human melanoma progression model and key insights into the transformation and heterogeneity of McSC-derived melanoma.


Assuntos
Carcinogênese/patologia , Melanócitos/patologia , Melanoma/patologia , Células-Tronco Neoplásicas/patologia , Animais , Carcinogênese/metabolismo , Transformação Celular Neoplásica/patologia , Derme/patologia , Modelos Animais de Doenças , Epiderme/patologia , Homeostase , Humanos , Melanócitos/metabolismo , Camundongos , Mutação/genética , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Microambiente Tumoral , Via de Sinalização Wnt
3.
Nat Commun ; 10(1): 1703, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979871

RESUMO

Multiple vertebrate embryonic structures such as organ primordia are composed of confluent cells. Although mechanisms that shape tissue sheets are increasingly understood, those which shape a volume of cells remain obscure. Here we show that 3D mesenchymal cell intercalations are essential to shape the mandibular arch of the mouse embryo. Using a genetically encoded vinculin tension sensor that we knock-in to the mouse genome, we show that cortical force oscillations promote these intercalations. Genetic loss- and gain-of-function approaches show that Wnt5a functions as a spatial cue to coordinate cell polarity and cytoskeletal oscillation. These processes diminish tissue rigidity and help cells to overcome the energy barrier to intercalation. YAP/TAZ and PIEZO1 serve as downstream effectors of Wnt5a-mediated actomyosin polarity and cytosolic calcium transients that orient and drive mesenchymal cell intercalations. These findings advance our understanding of how developmental pathways regulate biophysical properties and forces to shape a solid organ primordium.


Assuntos
Polaridade Celular , Citoesqueleto/fisiologia , Mandíbula/embriologia , Mandíbula/fisiologia , Proteína Wnt-5a/fisiologia , Citoesqueleto de Actina , Actomiosina/metabolismo , Animais , Cálcio/metabolismo , Ciclo Celular , Citosol/metabolismo , Elasticidade , Células Epiteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Mutação , Oscilometria , Transdução de Sinais , Estresse Mecânico , Vinculina/metabolismo , Viscosidade
4.
PLoS Genet ; 12(7): e1006150, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27414798

RESUMO

The Sonic hedgehog (Shh) signaling pathway regulates developmental, homeostatic, and repair processes throughout the body. In the skin, touch domes develop in tandem with primary hair follicles and contain sensory Merkel cells. The developmental signaling requirements for touch dome specification are largely unknown. We found dermal Wnt signaling and subsequent epidermal Eda/Edar signaling promoted Merkel cell morphogenesis by inducing Shh expression in early follicles. Lineage-specific gene deletions revealed intraepithelial Shh signaling was necessary for Merkel cell specification. Additionally, a Shh signaling agonist was sufficient to rescue Merkel cell differentiation in Edar-deficient skin. Moreover, Merkel cells formed in Fgf20 mutant skin where primary hair formation was defective but Shh production was preserved. Although developmentally associated with hair follicles, fate mapping demonstrated Merkel cells primarily originated outside the hair follicle lineage. These findings suggest that touch dome development requires Wnt-dependent mesenchymal signals to establish reciprocal signaling within the developing ectoderm, including Eda signaling to primary hair placodes and ultimately Shh signaling from primary follicles to extrafollicular Merkel cell progenitors. Shh signaling often demonstrates pleiotropic effects within a structure over time. In postnatal skin, Shh is known to regulate the self-renewal, but not the differentiation, of touch dome stem cells. Our findings relate the varied effects of Shh in the touch dome to the ligand source, with locally produced Shh acting as a morphogen essential for lineage specification during development and neural Shh regulating postnatal touch dome stem cell maintenance.


Assuntos
Ectodisplasinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Células de Merkel/citologia , Proteína Wnt1/metabolismo , Animais , Linhagem da Célula , Reparo do DNA , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Deleção de Genes , Genótipo , Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Homeostase , Ligantes , Masculino , Camundongos , Microscopia de Fluorescência , Morfogênese , Mutação , Neurônios/metabolismo , Transdução de Sinais , Pele/embriologia , Pele/metabolismo , Tato
5.
J Neurosci ; 35(17): 6836-48, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25926459

RESUMO

The ERK/MAPK pathway is an important developmental signaling pathway. Mutations in upstream elements of this pathway result in neuro-cardio-facial cutaneous (NCFC) syndromes, which are typified by impaired neurocognitive abilities that are reliant upon hippocampal function. The role of ERK signaling during hippocampal development has not been examined and may provide critical insight into the cause of hippocampal dysfunction in NCFC syndromes. In this study, we have generated ERK1 and conditional ERK2 compound knock-out mice to determine the role of ERK signaling during development of the hippocampal dentate gyrus. We found that loss of both ERK1 and ERK2 resulted in 60% fewer granule cells and near complete absence of neural progenitor pools in the postnatal dentate gyrus. Loss of ERK1/2 impaired maintenance of neural progenitors as they migrate from the dentate ventricular zone to the dentate gyrus proper, resulting in premature depletion of neural progenitor cells beginning at E16.5, which prevented generation of granule cells later in development. Finally, loss of ERK2 alone does not impair development of the dentate gyrus as animals expressing only ERK1 developed a normal hippocampus. These findings establish that ERK signaling regulates maintenance of progenitor cells required for development of the dentate gyrus.


Assuntos
Giro Denteado , Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Giro Denteado/embriologia , Giro Denteado/enzimologia , Giro Denteado/crescimento & desenvolvimento , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Ventrículos Laterais/citologia , Ventrículos Laterais/embriologia , Ventrículos Laterais/crescimento & desenvolvimento , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo
6.
Matrix Biol ; 31(7-8): 398-411, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23010571

RESUMO

ADAMTS-like proteins are related to ADAMTS metalloproteases by their similarity to ADAMTS ancillary domains. Here, we have characterized ADAMTSL5, a novel member of the superfamily with a unique modular organization that includes a single C-terminal netrin-like (NTR) module. Alternative splicing of ADAMTSL5 at its 5' end generates two transcripts that encode different signal peptides, but the same mature protein. These transcripts differ in their translational efficiency. Recombinant ADAMTSL5 is a secreted, N-glycosylated 60kDa glycoprotein located in the subcellular matrix, on the cell-surface, and in the medium of transfected cells. RT-PCR and western blot analysis of adult mouse tissues showed broad expression. Western blot analysis suggested proteolytic release of the NTR module in transfected cells as well as in some mouse tissues. Immunostaining during mouse organogenesis identified ADAMTSL5 in musculoskeletal tissues such as skeletal muscle, cartilage and bone, as well as in many epithelia. Affinity-chromatography demonstrated heparin-binding of ADAMTSL5 through its NTR-module. Recombinant ADAMTSL5 bound to both fibrillin-1 and fibrillin-2, and co-localized with fibrillin microfibrils in the extracellular matrix of cultured fibroblasts, but without discernible effect on microfibril assembly. ADAMTSL5 is the first family member shown to bind both fibrillin-1 and fibrillin-2. Like other ADAMTS proteins implicated in microfibril biology through identification of human and animal mutations, ADAMTSL5 could have a role in modulating microfibril functions.


Assuntos
Proteínas ADAM/metabolismo , Heparina/metabolismo , Microfibrilas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Recombinantes/metabolismo , Trombospondina 1/metabolismo , Proteínas ADAM/genética , Proteínas ADAMTS , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Imunofluorescência , Células HEK293 , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Netrina-1 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Trombospondina 1/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
7.
Arthritis Rheum ; 64(8): 2734-45, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22328118

RESUMO

OBJECTIVE: Fibrosis in human diseases and animal models is associated with aberrant Wnt/ß-catenin pathway activation. The aim of this study was to characterize the regulation, activity, mechanism of action, and significance of Wnt/ß-catenin signaling in the context of systemic sclerosis (SSc). METHODS: The expression of Wnt signaling pathway components in SSc skin biopsy specimens was analyzed. The regulation of profibrotic responses by canonical Wnt/ß-catenin was examined in explanted human mesenchymal cells. Fibrotic responses were studied using proliferation, migration, and gel contraction assays. The cell fate specification of subcutaneous preadipocytes by canonical Wnt signaling was evaluated. RESULTS: Analysis of published genome-wide expression data revealed elevated expression of the Wnt receptor FZD2 and the Wnt target LEF1 and decreased expression of Wnt antagonists DKK2 and WIF1 in skin biopsy specimens from subsets of patients with diffuse cutaneous SSc compared to the other distinct subsets. Immunohistochemical analysis showed increased nuclear ß-catenin expression in these biopsy specimens. In vitro, Wnt-3a induced ß-catenin activation, stimulated fibroblast proliferation and migration, collagen gel contraction, and myofibroblast differentiation, and enhanced profibrotic gene expression. Genetic and pharmacologic approaches were used to demonstrate that these profibrotic responses involved autocrine transforming growth factor ß signaling via Smads. In contrast, in explanted subcutaneous preadipocytes, Wnt-3a repressed adipogenesis and promoted myofibroblast differentiation. CONCLUSION: Canonical Wnt signaling was hyperactivated in SSc skin biopsy specimens. In explanted mesenchymal cells, Wnt-3a stimulated fibrogenic responses while suppressing adipogenesis. Taken together, these results indicate that Wnts have potent profibrotic effects, and that canonical Wnt signaling plays an important role in the pathogenesis of fibrosis and lipoatrophy in SSc.


Assuntos
Mesoderma/metabolismo , Mesoderma/patologia , Escleroderma Sistêmico/metabolismo , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Biópsia , Estudos de Casos e Controles , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Receptores Frizzled/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Mesoderma/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Escleroderma Sistêmico/patologia , Pele/metabolismo , Pele/patologia , Proteína Wnt3A/farmacologia
8.
Int J Biochem Cell Biol ; 40(12): 2720-38, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18620888

RESUMO

Mesenchymal cells are natural tissue builders. They exhibit an extraordinary capacity to metamorphize into differentiated cells, using extrinsic spatial and temporal inputs and intrinsic algorithms, as well as to build and adapt their own habitat. In addition to providing a habitat for osteoprogenitor cells, tissues of the skeletal system provide mechanical support and protection for the multiple organs of vertebrate organisms. This review examines the role of mechanics on determination of cell fate during pre-, peri- and postnatal development of the skeleton as well as during tissue genesis and repair in postnatal life. The role of cell mechanics is examined and brought into context of intrinsic cues during mesenchymal condensation. Remarkable new insights regarding structure function relationships in mesenchymal stem cells, and their influence on determination of cell fate are integrated in the context of de novo tissue generation and postnatal repair. Key differences in the formation of osteogenic and chondrogenic condensations are discussed in relation to direct intramembranous and indirect endochondral ossification. New approaches are discussed to elucidate and exploit extrinsic cues to generate tissues in the laboratory and in the clinic.


Assuntos
Fenômenos Fisiológicos Celulares , Mecanotransdução Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Diferenciação Celular , Linhagem da Célula , Previsões , Modelos Biológicos , Osteoblastos/citologia , Osteócitos/citologia , Osteogênese/genética , Engenharia Tecidual
9.
Dev Cell ; 4(2): 231-40, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12586066

RESUMO

Feather buds form sequentially in a hexagonal array. Bone morphogenetic protein (BMP) signaling from the feather bud inhibits bud formation in the adjacent interbud tissue, but whether interbud fate and patterning is actively promoted by BMP or other factors is unclear. We show that epidermal growth factor (EGF) signaling acts positively to establish interbud identity. EGF and the active EGF receptor (EGFR) are expressed in the interbud regions. Exogenous EGF stimulates epidermal proliferation and expands interbud gene expression, with a concurrent loss of feather bud gene expression and morphology. Conversely, EGFR inhibitors result in the loss of interbud fate and increased acquisition of feather bud fate. EGF signaling acts directly on the epidermis and is independent of BMP signaling. The timing of competence to interpret interbud-promoting signals occurs at an earlier developmental stage than previously anticipated. These data demonstrate that EGFR signaling actively promotes interbud identity.


Assuntos
Fator de Crescimento Epidérmico/fisiologia , Plumas/embriologia , Transdução de Sinais/fisiologia , Pele/metabolismo , Animais , Padronização Corporal , Proteínas Morfogenéticas Ósseas/fisiologia , Bromodesoxiuridina , Divisão Celular , Embrião de Galinha , Galinhas , Técnicas de Cultura , Fator de Crescimento Epidérmico/farmacologia , Epiderme , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Plumas/citologia , Plumas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas Imunoenzimáticas , Hibridização In Situ , Pele/citologia
10.
J Neurosci ; 22(22): 9831-40, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12427839

RESUMO

The neural crest gives rise to numerous cell types, including Schwann cells, neurons, and melanocytes. The extent to which adult neural crest-derived cells retain plasticity has not been tested previously. We report that cutting adult mouse sciatic nerve induces pigmentation around nerve fascicles, among muscle bundles, and in the hypodermis. Pigmented cells are derived from adult nerve, because pigmentation occurs even when nerve fragments are grafted into tyrosinase null albino mice. Pigmentation defects are pervasive in patients with neurofibromatosis type 1 (NF1). Mice hemizygous for Nf1 mutations show enhanced pigmentation after nerve lesion and occasionally form pigmented and unpigmented tumors. The Nf1 nerve and the Nf1 host environment both contribute to enhanced pigmentation. Grafted purified Nf1 mutant glial cells [S100(+)-p75NGFR(+)-GFAP(+)-EGFR(+) or S100(+)-p75NGFR(+)-GFAP(+)-EGFR(-)] mimic nerve-derived pigmentation. The NF1 protein, neurofibromin, is a Ras-GAP that acts downstream of a few defined receptor tyrosine kinases, including [beta-common (beta(c))] the shared common receptor for granulocyte and monocyte colony-stimulating factor, interleukin-3 (IL3), and IL5. Cytokines in the environment have the potential to suppress pigmentation as shown by nerve injury experiments in null mice; when is beta(c) absent or Nf1 is mutant, melanogenesis is increased. Thus, the adult nerve glial cell phenotype is maintained after nerve injury by response to cytokines, through neurofibromin.


Assuntos
Citocinas/metabolismo , Melaninas/biossíntese , Neuroglia/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Neuropatia Ciática/fisiopatologia , Transdução de Sinais/fisiologia , Animais , Axotomia , Diferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/transplante , Heterozigoto , Masculino , Melanócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Compressão Nervosa , Crista Neural/citologia , Crista Neural/embriologia , Neurofibromina 1/deficiência , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Neuroglia/citologia , Neoplasias do Sistema Nervoso Periférico/complicações , Pigmentação , Células de Schwann/citologia , Células de Schwann/metabolismo , Células de Schwann/transplante , Nervo Isquiático/lesões , Nervo Isquiático/transplante , Neuropatia Ciática/complicações , Neuropatia Ciática/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA