Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biochem Biophys Res Commun ; 596: 29-35, 2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-35108651

RESUMO

Thymic dendritic cells (DCs) promote immune tolerance by regulating negative selection of autoreactive T cells in the thymus. How DC homing to the thymus is transcriptionally regulated is still unclear. Microphthalmia-associated transcription factor (Mitf) is broadly expressed and plays essential roles in the hematopoietic system. Here, we used Mitf-mutated mice (Mitfvit/vit) and found enlargement of the thymus and expansion of CD4/CD8 double-positive T cells. Mitf was highly expressed in a subset of thymic DCs among the hematopoietic system. Genetic mutation or pharmacological inhibition of Mitf in DCs decreased the expression levels of Itga4, which are critical molecules for the homing of DCs to the thymus. Further, inhibition of Mitf decreased thymic DC number. These results suggest a pivotal role of Mitf in the maintenance of T cell differentiation by regulating the homing of DC subsets within the thymus.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Fator de Transcrição Associado à Microftalmia/imunologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Células Dendríticas/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Hiperplasia , Integrina alfa4/genética , Integrina alfa4/imunologia , Integrina alfa4/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Timo/metabolismo , Timo/patologia
2.
J Cell Biol ; 220(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34550317

RESUMO

Skin regenerative capacity declines with age, but the underlying mechanisms are largely unknown. Here we demonstrate a functional link between epidermal growth factor receptor (EGFR) signaling and type XVII collagen (COL17A1) proteolysis on age-associated alteration of keratinocyte stem cell dynamics in skin regeneration. Live-imaging and computer simulation experiments predicted that human keratinocyte stem cell motility is coupled with self-renewal and epidermal regeneration. Receptor tyrosine kinase array identified the age-associated decline of EGFR signaling in mouse skin wound healing. Culture experiments proved that EGFR activation drives human keratinocyte stem cell motility with increase of COL17A1 by inhibiting its proteolysis through the secretion of tissue inhibitor of metalloproteinases 1 (TIMP1). Intriguingly, COL17A1 directly regulated keratinocyte stem cell motility and collective cell migration by coordinating actin and keratin filament networks. We conclude that EGFR-COL17A1 axis-mediated keratinocyte stem cell motility drives epidermal regeneration, which provides a novel therapeutic approach for age-associated impaired skin regeneration.


Assuntos
Autoantígenos/metabolismo , Movimento Celular/fisiologia , Colágenos não Fibrilares/metabolismo , Regeneração/fisiologia , Pele/metabolismo , Células 3T3 , Animais , Linhagem Celular , Células Epidérmicas/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteólise , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Cicatrização/fisiologia , Colágeno Tipo XVII
3.
Cell Rep ; 36(5): 109492, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348144

RESUMO

Early differential diagnosis between malignant and benign tumors and their underlying intrinsic differences are the most critical issues for life-threatening cancers. To study whether human acral melanomas, deadly cancers that occur on non-hair-bearing skin, have distinct origins that underlie their invasive capability, we develop fate-tracing technologies of melanocyte stem cells in sweat glands (glandular McSCs) and in melanoma models in mice and compare the cellular dynamics with human melanoma. Herein, we report that glandular McSCs self-renew to expand their migratory progeny in response to genotoxic stress and trauma to generate invasive melanomas in mice that mimic human acral melanomas. The analysis of melanocytic lesions in human volar skin reveals that genetically unstable McSCs expand in sweat glands and in the surrounding epidermis in melanomas but not in nevi. The detection of such cell spreading dynamics provides an innovative method for an early differential diagnosis of acral melanomas from nevi.


Assuntos
Movimento Celular , Melanoma/patologia , Nevo/patologia , Células-Tronco/patologia , Animais , Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Ciclina D1/metabolismo , Modelos Animais de Doenças , Epiderme/patologia , Epiderme/efeitos da radiação , Amplificação de Genes , Instabilidade Genômica/efeitos da radiação , Melanócitos/patologia , Melanócitos/efeitos da radiação , Melanoma/diagnóstico , Camundongos Endogâmicos C57BL , Fatores de Risco , Pele/patologia , Pele/efeitos da radiação , Pigmentação da Pele/efeitos da radiação , Glândulas Sudoríparas/efeitos da radiação , Raios Ultravioleta
4.
Nat Commun ; 12(1): 1826, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758188

RESUMO

Somatic mutations of ASXL1 are frequently detected in age-related clonal hematopoiesis (CH). However, how ASXL1 mutations drive CH remains elusive. Using knockin (KI) mice expressing a C-terminally truncated form of ASXL1-mutant (ASXL1-MT), we examined the influence of ASXL1-MT on physiological aging in hematopoietic stem cells (HSCs). HSCs expressing ASXL1-MT display competitive disadvantage after transplantation. Nevertheless, in genetic mosaic mouse model, they acquire clonal advantage during aging, recapitulating CH in humans. Mechanistically, ASXL1-MT cooperates with BAP1 to deubiquitinate and activate AKT. Overactive Akt/mTOR signaling induced by ASXL1-MT results in aberrant proliferation and dysfunction of HSCs associated with age-related accumulation of DNA damage. Treatment with an mTOR inhibitor rapamycin ameliorates aberrant expansion of the HSC compartment as well as dysregulated hematopoiesis in aged ASXL1-MT KI mice. Our findings suggest that ASXL1-MT provokes dysfunction of HSCs, whereas it confers clonal advantage on HSCs over time, leading to the development of CH.


Assuntos
Envelhecimento/genética , Hematopoiese Clonal/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas Repressoras/genética , Serina-Treonina Quinases TOR/metabolismo , Idoso , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Animais , Apoptose/genética , Ciclo Celular/genética , Proliferação de Células/genética , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Técnicas de Introdução de Genes , Hematopoese/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA-Seq , Espécies Reativas de Oxigênio/farmacologia , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sirolimo/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/genética
5.
J Dermatol Sci ; 97(2): 143-151, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32001115

RESUMO

BACKGROUND: NUAK2 is a critical gene that participates in the carcinogenesis of various types of cancers including melanomas. However, the expression patterns of NUAK2 in normal skin and in various types of skin tumors have not been fully elucidated to date. OBJECTIVES: To elucidate the distribution and localization of NUAK2 expression in normal skin, and characterize the expression patterns of NUAK2 and YAP in various types of skin tumors. METHODS: In this study, we characterized the expression of NUAK2 in tissues by developing a novel NUAK2-specific monoclonal antibody and using that to determine NUAK2 expression patterns in normal skin and in 155 cases of various types of skin tumors, including extramammary Paget's disease (EMPD), squamous cell carcinoma (SCC), Bowen's disease (BD), actinic keratosis (AK), basal cell carcinoma (BCC) and angiosarcoma (AS). Further, we analyzed the expression patterns of YAP and p-Akt in those tumors. RESULTS: Our analyses revealed that NUAK2 is expressed at high frequencies in EMPD, SCC, BD, AK, BCC and AS. The expression of p-Akt was positively correlated with tumor size in EMPD (P = 0.001). Importantly, the expression of NUAK2 was significantly correlated with YAP in SCC (P = 0.012) and in BD (P = 0.009). CONCLUSIONS: Our results suggest that the YAP-NUAK2 axis has critical importance in the tumorigenesis of SCC and BD, and that therapeutic modalities targeting the YAP-NUAK2 axis may be an effective approach against skin tumors including SCC and BD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinogênese/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Bowen/patologia , Carcinogênese/genética , Carcinoma de Células Escamosas/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Serina-Treonina Quinases/análise , Estudos Retrospectivos , Transdução de Sinais/genética , Pele/metabolismo , Pele/patologia , Fatores de Transcrição/análise , Proteínas de Sinalização YAP
6.
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
7.
Cell Stem Cell ; 19(2): 192-204, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27345838

RESUMO

Hematopoietic stem cells (HSCs) maintain quiescence by activating specific metabolic pathways, including glycolysis. We do not yet have a clear understanding of how this metabolic activity changes during stress hematopoiesis, such as bone marrow transplantation. Here, we report a critical role for the p38MAPK family isoform p38α in initiating hematopoietic stem and progenitor cell (HSPC) proliferation during stress hematopoiesis in mice. We found that p38MAPK is immediately phosphorylated in HSPCs after a hematological stress, preceding increased HSPC cycling. Conditional deletion of p38α led to defective recovery from hematological stress and a delay in initiation of HSPC proliferation. Mechanistically, p38α signaling increases expression of inosine-5'-monophosphate dehydrogenase 2 in HSPCs, leading to altered levels of amino acids and purine-related metabolites and changes in cell-cycle progression in vitro and in vivo. Our studies have therefore uncovered a p38α-mediated pathway that alters HSPC metabolism to respond to stress and promote recovery.


Assuntos
Ciclo Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Purinas/metabolismo , Estresse Fisiológico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose , Proliferação de Células , Ativação Enzimática , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/enzimologia , IMP Desidrogenase/antagonistas & inibidores , IMP Desidrogenase/metabolismo , Camundongos Endogâmicos C57BL , Fator de Transcrição Associado à Microftalmia/metabolismo , Mutação/genética , Fenótipo , Espécies Reativas de Oxigênio/metabolismo
8.
Pigment Cell Melanoma Res ; 27(6): 1039-50, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25065272

RESUMO

Determination of the niche for early-stage cancer remains a challenging issue. Melanoma is an aggressive cancer of the melanocyte lineage. Early melanoma cells are often found in the epidermis around sweat ducts of human volar skin, and the skin pigmentation pattern is an early diagnostic sign of acral melanoma. However, the niche for melanoma precursors has not been determined yet. Here, we report that the secretory portion (SP) of eccrine sweat glands provide an anatomical niche for melanocyte-melanoma precursor cells. Using lineage-tagged H2B-GFP reporter mice, we found that melanoblasts that colonize sweat glands during development are maintained in an immature, slow-cycling state but renew themselves in response to genomic stress and provide their differentiating progeny to the epidermis. FISH analysis of human acral melanoma expanding in the epidermis revealed that unpigmented melanoblasts with significant cyclin D1 gene amplification reside deep in the SP of particular sweat gland(s). These findings indicate that sweat glands maintain melanocyte-melanoma precursors in an immature state in the niche and explain the preferential distribution of early melanoma cells around sweat glands in human volar skin.


Assuntos
Melanócitos/patologia , Melanoma/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Cutâneas/patologia , Pele/patologia , Nicho de Células-Tronco , Glândulas Sudoríparas/patologia , Animais , Ciclo Celular , Ciclina D1/genética , Amplificação de Genes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos
10.
Pigment Cell Melanoma Res ; 27(4): 540-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24730534

RESUMO

Current studies have revealed that stem cells are more radiosensitive than mature cells. As somatic stem cells are mostly kept in a quiescent state, this conflicts with Bergonié and Tribondeau's law that actively mitotic cells are the most radiosensitive. In this study, we focused on hair graying to understand the stress-resistance of melanocyte stem cells (McSCs). We used Dct-H2B-GFP transgenic mice which enables the stable visualization of McSCs and an anti-Kit monoclonal antibody which selectively eradicates amplifying McSCs. The results demonstrate that quiescent McSCs are rather radiosensitive, but the coexistence of non-quiescent McSCs provides the stem cell pool with radioresistance. The irradiated quiescent McSCs prematurely differentiate in the niche upon their activation without sufficiently renewing themselves for cyclic hair pigmentation. These data indicate that tissue radiosensitivity is largely dependent on the state of somatic stem cells under their local microenvironment.


Assuntos
Folículo Piloso/metabolismo , Melanócitos/metabolismo , Pigmentação/efeitos da radiação , Tolerância a Radiação/efeitos da radiação , Nicho de Células-Tronco/efeitos da radiação , Células-Tronco/metabolismo , Animais , Folículo Piloso/patologia , Melanócitos/patologia , Camundongos , Camundongos Transgênicos , Pigmentação/genética , Tolerância a Radiação/genética , Nicho de Células-Tronco/genética , Células-Tronco/patologia , Raios X
11.
Int J Radiat Biol ; 88(6): 501-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22489999

RESUMO

PURPOSE: New developments in knowledge of radiation effects on tissue stem cells were discussed in a Workshop held at the Radiation Effects Research Foundation (RERF) in Hiroshima, Japan, 18-19 January 2012. RESULTS: Stem cells and their niche in intestinal mucosa, haemopoietic tissue, hair follicles, and spermatogenesis were discussed variously with regard to radiosensitivity, repair, regeneration, age-dependency of effects, genetic effects, and protection aspects. These tissues all possess a common basic template, but there are structural and hierarchical differences between tissues which continue to be elucidated in terms of a stem-cell age structure and niche regulatory signals which together govern radiation responses. CONCLUSIONS: Stem cells and their niche have become much better characterized in recent years, and their radiation response can be elucidated in detail in experimental systems to help underpin both protection and therapeutic recommendations established from human epidemiological evidence. This report summarizes the presentations at the meeting, and concludes with some remaining questions which may be answered with the help of this type of research.


Assuntos
Mutação , Células-Tronco/efeitos da radiação , Animais , Mutação em Linhagem Germinativa , Humanos , Masculino , Lesões por Radiação/genética , Tolerância a Radiação , Regeneração , Espermatogênese/genética , Espermatogênese/efeitos da radiação , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/fisiologia , Nicho de Células-Tronco/efeitos da radiação
12.
Pigment Cell Melanoma Res ; 24(3): 401-10, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21466661

RESUMO

Most mammals are coated with pigmented hair. Melanocytes in each hair follicle produce melanin pigments for the hair during each hair cycle. The key to understanding the mechanism of cyclic melanin production is the melanocyte stem cell (MelSC) population, previously known as 'amelanotic melanocytes'. The MelSCs directly adhere to hair follicle stem cells, the niche cells for MelSCs and reside in the hair follicle bulge-subbulge area, the lower permanent portion of the hair follicle, to serve as a melanocyte reservoir for skin and hair pigmentation. MelSCs form a stem cell system within individual hair follicles and provide a 'hair pigmentary unit' for each cycle of hair pigmentation. This review focuses on the identification of MelSCs and their characteristics and explains the importance of the MelSC population in the mechanisms of hair pigmentation, hair greying, and skin repigmentation.


Assuntos
Folículo Piloso/metabolismo , Melaninas/metabolismo , Melanócitos/metabolismo , Pigmentação da Pele/fisiologia , Células-Tronco/metabolismo , Animais , Folículo Piloso/citologia , Humanos , Melanócitos/citologia , Camundongos , Células-Tronco/citologia
13.
Cell Stem Cell ; 6(2): 130-40, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-20144786

RESUMO

Melanocyte stem cells in the bulge area of hair follicles are responsible for hair pigmentation, and defects in them cause hair graying. Here we describe the process of melanocyte stem cell entry into the quiescent state and show that niche-derived transforming growth factor beta (TGF-beta) signaling plays important roles in this process. In vitro, TGF-beta not only induces reversible cell cycle arrest, but also promotes melanocyte immaturity by downregulating MITF, the master transcriptional regulator of melanocyte differentiation, and its downstream melanogenic genes. In vivo, TGF-beta signaling is activated in melanocyte stem cells when they reenter the quiescent noncycling state during the hair cycle and this process requires Bcl2 for cell survival. Furthermore, targeted TGF-beta type II receptor (TGFbRII) deficiency in the melanocyte lineage causes incomplete maintenance of melanocyte stem cell immaturity and results in mild hair graying. These data demonstrate that the TGF-beta signaling pathway is one of the key niche factors that regulate melanocyte stem cell immaturity and quiescence.


Assuntos
Melanócitos/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Humanos , Melanócitos/citologia , Melanócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Células-Tronco/citologia , Células-Tronco/imunologia , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta2/imunologia , Fator de Crescimento Transformador beta3/imunologia
14.
Cell ; 137(6): 1088-99, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19524511

RESUMO

Somatic stem cell depletion due to the accumulation of DNA damage has been implicated in the appearance of aging-related phenotypes. Hair graying, a typical sign of aging in mammals, is caused by the incomplete maintenance of melanocyte stem cells (MSCs) with age. Here, we report that irreparable DNA damage, as caused by ionizing radiation, abrogates renewal of MSCs in mice. Surprisingly, the DNA-damage response triggers MSC differentiation into mature melanocytes in the niche, rather than inducing their apoptosis or senescence. The resulting MSC depletion leads to irreversible hair graying. Furthermore, deficiency of Ataxia-telangiectasia mutated (ATM), a central transducer kinase of the DNA-damage response, sensitizes MSCs to ectopic differentiation, demonstrating that the kinase protects MSCs from their premature differentiation by functioning as a "stemness checkpoint" to maintain the stem cell quality and quantity.


Assuntos
Diferenciação Celular , Dano ao DNA , Melanócitos/citologia , Melanócitos/efeitos da radiação , Células-Tronco/citologia , Células-Tronco/efeitos da radiação , Envelhecimento , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Cabelo/citologia , Cabelo/patologia , Cabelo/fisiopatologia , Melanossomas/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Raios X
15.
Mol Cell ; 32(4): 554-63, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-19026785

RESUMO

The protein encoded by paired-box homeotic gene 3 (PAX3) is a key regulator of the microphthalmia-associated transcription factor (Mitf) in the melanocyte lineage. Here, we show that PAX3 expression in skin is directly inhibited by TGF-beta/Smads. UV irradiation represses TGF-beta in keratinocytes, and the repression of TGF-beta/Smads upregulates PAX3 in melanocytes, which is associated with a UV-induced melanogenic response and consequent pigmentation. Furthermore, the TGF-beta-PAX3 signaling pathway interacts with the p53-POMC/MSH-MC1R signaling pathway, and both are crucial in melanogenesis. The activation of p53-POMC/MSH-MC1R signaling is required for the UV-induced melanogenic response because PAX3 functions in synergy with SOX10 in a cAMP-response element (CRE)-dependent manner to regulate the transcription of Mitf. This study will provide a rich foundation for further research on skin cancer prevention by enabling us to identify targeted small molecules in the signaling pathways of the UV-induced melanogenic response that are highly likely to induce naturally protective pigmentation.


Assuntos
Regulação da Expressão Gênica , Melanócitos/fisiologia , Fatores de Transcrição Box Pareados/antagonistas & inibidores , Fatores de Transcrição Box Pareados/genética , Fator de Crescimento Transformador beta/metabolismo , Genes Reguladores , Genes Reporter , Humanos , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Luciferases/metabolismo , Melanócitos/metabolismo , Fator de Transcrição PAX3 , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Pigmentação da Pele/genética , Pigmentação da Pele/fisiologia , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/genética , Raios Ultravioleta
16.
Nature ; 443(7109): 340-4, 2006 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16988713

RESUMO

Ultraviolet-light (UV)-induced tanning is defective in numerous 'fair-skinned' individuals, many of whom contain functional disruption of the melanocortin 1 receptor (MC1R). Although this suggested a critical role for the MC1R ligand melanocyte stimulating hormone (MSH) in this response, a genetically controlled system has been lacking in which to determine the precise role of MSH-MC1R. Here we show that ultraviolet light potently induces expression of MSH in keratinocytes, but fails to stimulate pigmentation in the absence of functional MC1R in red/blonde-haired Mc1r(e/e) mice. However, pigmentation could be rescued by topical application of the cyclic AMP agonist forskolin, without the need for ultraviolet light, demonstrating that the pigmentation machinery is available despite the absence of functional MC1R. This chemically induced pigmentation was protective against ultraviolet-light-induced cutaneous DNA damage and tumorigenesis when tested in the cancer-prone, xeroderma-pigmentosum-complementation-group-C-deficient genetic background. These data emphasize the essential role of intercellular MSH signalling in the tanning response, and suggest a clinical strategy for topical small-molecule manipulation of pigmentation.


Assuntos
Colforsina/administração & dosagem , Colforsina/farmacologia , Receptor Tipo 1 de Melanocortina/metabolismo , Dermatopatias/prevenção & controle , Pigmentação da Pele/efeitos dos fármacos , Pigmentação da Pele/efeitos da radiação , Raios Ultravioleta , Administração Tópica , Animais , Melaninas/biossíntese , Hormônios Estimuladores de Melanócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor Tipo 1 de Melanocortina/deficiência , Receptor Tipo 1 de Melanocortina/genética , Transdução de Sinais , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Pele/efeitos da radiação , Dermatopatias/etiologia , Dermatopatias/patologia , Pigmentação da Pele/fisiologia
17.
J Biol Chem ; 281(15): 10365-73, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16455654

RESUMO

Hepatocyte growth factor (HGF)/c-Met signaling is thought to be a key pathway in both melanocyte development and melanoma metastasis. Here, HGF stimulation of melanocytes was seen to up-regulate c-Met expression. In an effort to decipher the mechanism by which HGF up-regulates its receptor, we found that c-Met is a direct transcriptional target of Mitf. This was confirmed with chromatin immunoprecipitation experiments of the human c-Met promoter, as well as by the ability of adenovirally expressed Mitf to modulate endogenous c-Met protein levels in melanocytes. Disruption of Mitf blocked HGF-dependent increases in endogenous c-Met message and protein levels, indicating that HGF regulates its own receptor levels via Mitf. Finally, dominant-negative inhibition of Mitf resulted in profound resistance of melanocytes and melanoma cells to HGF-dependent matrix invasion, suggesting a physiologic role for this pathway in melanocytic development and melanoma.


Assuntos
Melanócitos/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas Proto-Oncogênicas c-met/química , Adenoviridae/genética , Adenoviridae/metabolismo , Western Blotting , Linhagem Celular Tumoral , Linhagem da Célula , Núcleo Celular/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Primers do DNA/química , Regulação da Expressão Gênica , Genes Dominantes , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Immunoblotting , Cinética , Sistema de Sinalização das MAP Quinases , Melanoma/metabolismo , Melanoma/patologia , Modelos Genéticos , Metástase Neoplásica , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-met/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transcrição Gênica , Regulação para Cima
18.
Science ; 307(5710): 720-4, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15618488

RESUMO

Hair graying is the most obvious sign of aging in humans, yet its mechanism is largely unknown. Here, we used melanocyte-tagged transgenic mice and aging human hair follicles to demonstrate that hair graying is caused by defective self-maintenance of melanocyte stem cells. This process is accelerated dramatically with Bcl2 deficiency, which causes selective apoptosis of melanocyte stem cells, but not of differentiated melanocytes, within the niche at their entry into the dormant state. Furthermore, physiologic aging of melanocyte stem cells was associated with ectopic pigmentation or differentiation within the niche, a process accelerated by mutation of the melanocyte master transcriptional regulator Mitf.


Assuntos
Envelhecimento/fisiologia , Cor de Cabelo , Folículo Piloso/fisiologia , Melanócitos/fisiologia , Células-Tronco/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose , Diferenciação Celular , Forma Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Folículo Piloso/citologia , Humanos , Melaninas/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição Associado à Microftalmia , Pessoa de Meia-Idade , Morfogênese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vibrissas/citologia , Vibrissas/fisiologia
19.
Cancer Cell ; 6(6): 565-76, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15607961

RESUMO

The genomic organization of the CDK2 gene, which overlaps the melanocyte-specific gene SILV/PMEL17, poses an interesting regulatory challenge. We show that, despite its ubiquitous expression, CDK2 exhibits tissue-specific regulation by the essential melanocyte lineage transcription factor MITF. In addition, functional studies revealed this regulation to be critical for maintaining CDK2 kinase activity and growth of melanoma cells. Expression levels of MITF and CDK2 are tightly correlated in primary melanoma specimens and predict susceptibility to the CDK2 inhibitor roscovitine. CDK2 depletion suppressed growth and cell cycle progression in melanoma, but not other cancers, corroborating previous results. Collectively, these data indicate that CDK2 activity in melanoma is largely maintained at the transcriptional level by MITF, and unlike other malignancies, it may be a suitable drug target in melanoma.


Assuntos
Quinases relacionadas a CDC2 e CDC28/fisiologia , Proteínas de Ligação a DNA/fisiologia , Melanoma/patologia , Fatores de Transcrição/fisiologia , Western Blotting , Quinases relacionadas a CDC2 e CDC28/genética , Quinases relacionadas a CDC2 e CDC28/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Imunoprecipitação da Cromatina , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Elementos E-Box/fisiologia , Fibroblastos/metabolismo , Citometria de Fluxo , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Reporter/genética , Humanos , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/metabolismo , Glicoproteínas de Membrana , Fator de Transcrição Associado à Microftalmia , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Inibidores de Proteínas Quinases/farmacologia , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Purinas/farmacologia , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Roscovitina , Fase S/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Proteína bcl-X , Antígeno gp100 de Melanoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA