Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 35(23-24): 1657-1677, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34819350

RESUMEN

Senescence shapes embryonic development, plays a key role in aging, and is a critical barrier to cancer initiation, yet how senescence is regulated remains incompletely understood. TBX2 is an antisenescence T-box family transcription repressor implicated in embryonic development and cancer. However, the repertoire of TBX2 target genes, its cooperating partners, and how TBX2 promotes proliferation and senescence bypass are poorly understood. Here, using melanoma as a model, we show that TBX2 lies downstream from PI3K signaling and that TBX2 binds and is required for expression of E2F1, a key antisenescence cell cycle regulator. Remarkably, TBX2 binding in vivo is associated with CACGTG E-boxes, present in genes down-regulated by TBX2 depletion, more frequently than the consensus T-element DNA binding motif that is restricted to Tbx2 repressed genes. TBX2 is revealed to interact with a wide range of transcription factors and cofactors, including key components of the BCOR/PRC1.1 complex that are recruited by TBX2 to the E2F1 locus. Our results provide key insights into how PI3K signaling modulates TBX2 function in cancer to drive proliferation.


Asunto(s)
Melanoma , Proteínas de Dominio T Box , Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo
2.
Cell ; 155(5): 1022-33, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267888

RESUMEN

Sequence polymorphisms linked to human diseases and phenotypes in genome-wide association studies often affect noncoding regions. A SNP within an intron of the gene encoding Interferon Regulatory Factor 4 (IRF4), a transcription factor with no known role in melanocyte biology, is strongly associated with sensitivity of skin to sun exposure, freckles, blue eyes, and brown hair color. Here, we demonstrate that this SNP lies within an enhancer of IRF4 transcription in melanocytes. The allele associated with this pigmentation phenotype impairs binding of the TFAP2A transcription factor that, together with the melanocyte master regulator MITF, regulates activity of the enhancer. Assays in zebrafish and mice reveal that IRF4 cooperates with MITF to activate expression of Tyrosinase (TYR), an essential enzyme in melanin synthesis. Our findings provide a clear example of a noncoding polymorphism that affects a phenotype by modulating a developmental gene regulatory network.


Asunto(s)
Factores Reguladores del Interferón/metabolismo , Polimorfismo de Nucleótido Simple , Animales , Secuencia de Bases , Elementos de Facilitación Genéticos , Humanos , Factores Reguladores del Interferón/química , Factores Reguladores del Interferón/genética , Melanocitos/metabolismo , Ratones , Datos de Secuencia Molecular , Pigmentación , Transducción de Señal , Factor de Transcripción AP-2/química , Factor de Transcripción AP-2/metabolismo , Pez Cebra
3.
Genes Dev ; 33(5-6): 310-332, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30804224

RESUMEN

Whether cell types exposed to a high level of environmental insults possess cell type-specific prosurvival mechanisms or enhanced DNA damage repair capacity is not well understood. BRN2 is a tissue-restricted POU domain transcription factor implicated in neural development and several cancers. In melanoma, BRN2 plays a key role in promoting invasion and regulating proliferation. Here we found, surprisingly, that rather than interacting with transcription cofactors, BRN2 is instead associated with DNA damage response proteins and directly binds PARP1 and Ku70/Ku80. Rapid PARP1-dependent BRN2 association with sites of DNA damage facilitates recruitment of Ku80 and reprograms DNA damage repair by promoting Ku-dependent nonhomologous end-joining (NHEJ) at the expense of homologous recombination. BRN2 also suppresses an apoptosis-associated gene expression program to protect against UVB-, chemotherapy- and vemurafenib-induced apoptosis. Remarkably, BRN2 expression also correlates with a high single-nucleotide variation prevalence in human melanomas. By promoting error-prone DNA damage repair via NHEJ and suppressing apoptosis of damaged cells, our results suggest that BRN2 contributes to the generation of melanomas with a high mutation burden. Our findings highlight a novel role for a key transcription factor in reprogramming DNA damage repair and suggest that BRN2 may impact the response to DNA-damaging agents in BRN2-expressing cancers.


Asunto(s)
Apoptosis , Reparación del ADN por Unión de Extremidades/genética , Proteínas de Homeodominio/metabolismo , Melanoma/genética , Melanoma/fisiopatología , Mutación/genética , Factores del Dominio POU/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Humanos , Autoantígeno Ku/metabolismo , Factores del Dominio POU/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica , Dominios Proteicos , Transporte de Proteínas
4.
Cell ; 146(1): 67-79, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21722948

RESUMEN

DNA methylation is a major epigenetic mechanism for gene silencing. Whereas methyltransferases mediate cytosine methylation, it is less clear how unmethylated regions in mammalian genomes are protected from de novo methylation and whether an active demethylating activity is involved. Here, we show that either knockout or catalytic inactivation of the DNA repair enzyme thymine DNA glycosylase (TDG) leads to embryonic lethality in mice. TDG is necessary for recruiting p300 to retinoic acid (RA)-regulated promoters, protection of CpG islands from hypermethylation, and active demethylation of tissue-specific developmentally and hormonally regulated promoters and enhancers. TDG interacts with the deaminase AID and the damage response protein GADD45a. These findings highlight a dual role for TDG in promoting proper epigenetic states during development and suggest a two-step mechanism for DNA demethylation in mammals, whereby 5-methylcytosine and 5-hydroxymethylcytosine are first deaminated by AID to thymine and 5-hydroxymethyluracil, respectively, followed by TDG-mediated thymine and 5-hydroxymethyluracil excision repair.


Asunto(s)
Metilación de ADN , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Timina ADN Glicosilasa/metabolismo , 5-Metilcitosina/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Citidina Desaminasa/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Timina ADN Glicosilasa/genética , Transcripción Genética
5.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34878101

RESUMEN

The canonical Wnt/ß-catenin pathway governs a multitude of developmental processes in various cell lineages, including the melanocyte lineage. Indeed, ß-catenin regulates transcription of Mitf-M, the master regulator of this lineage. The first wave of melanocytes to colonize the skin is directly derived from neural crest cells, whereas the second wave of melanocytes is derived from Schwann cell precursors (SCPs). We investigated the influence of ß-catenin in the development of melanocytes of the first and second waves by generating mice expressing a constitutively active form of ß-catenin in cells expressing tyrosinase. Constitutive activation of ß-catenin did not affect the development of truncal melanoblasts but led to marked hyperpigmentation of the paws. By activating ß-catenin at various stages of development (E8.5-E11.5), we showed that the activation of ß-catenin in bipotent SCPs favored melanoblast specification at the expense of Schwann cells in the limbs within a specific temporal window. Furthermore, in vitro hyperactivation of the Wnt/ß-catenin pathway, which is required for melanocyte development, induces activation of Mitf-M, in turn repressing FoxD3 expression. In conclusion, ß-catenin overexpression promotes SCP cell fate decisions towards the melanocyte lineage.


Asunto(s)
Diferenciación Celular , Melanocitos/metabolismo , Células de Schwann/citología , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Linaje de la Célula , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Melanocitos/citología , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Estabilidad Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Células de Schwann/metabolismo , Vía de Señalización Wnt , beta Catenina/genética
6.
Cell ; 141(6): 994-1005, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20550935

RESUMEN

DICER is a central regulator of microRNA maturation. However, little is known about mechanisms regulating its expression in development or disease. While profiling miRNA expression in differentiating melanocytes, two populations were observed: some upregulated at the pre-miRNA stage, and others upregulated as mature miRNAs (with stable pre-miRNA levels). Conversion of pre-miRNAs to fully processed miRNAs appeared to be dependent upon stimulation of DICER expression--an event found to occur via direct transcriptional targeting of DICER by the melanocyte master transcriptional regulator MITF. MITF binds and activates a conserved regulatory element upstream of DICER's transcriptional start site upon melanocyte differentiation. Targeted KO of DICER is lethal to melanocytes, at least partly via DICER-dependent processing of the pre-miRNA-17 approximately 92 cluster thus targeting BIM, a known proapoptotic regulator of melanocyte survival. These observations highlight a central mechanism underlying lineage-specific miRNA regulation which could exist for other cell types during development.


Asunto(s)
Regulación de la Expresión Génica , Melanocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Ribonucleasa III/metabolismo , Transcripción Genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Células Epidérmicas , Técnicas de Silenciamiento del Gen , Folículo Piloso/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Regulación hacia Arriba
7.
Development ; 145(12)2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29769218

RESUMEN

To distribute and establish the melanocyte lineage throughout the skin and other developing organs, melanoblasts undergo several rounds of proliferation, accompanied by migration through complex environments and differentiation. Melanoblast migration requires interaction with extracellular matrix of the epidermal basement membrane and with surrounding keratinocytes in the developing skin. Migration has been characterized by measuring speed, trajectory and directionality of movement, but there are many unanswered questions about what motivates and defines melanoblast migration. Here, we have established a general mathematical model to simulate the movement of melanoblasts in the epidermis based on biological data, assumptions and hypotheses. Comparisons between experimental data and computer simulations reinforce some biological assumptions, and suggest new ideas for how melanoblasts and keratinocytes might influence each other during development. For example, it appears that melanoblasts instruct each other to allow a homogeneous distribution in the tissue and that keratinocytes may attract melanoblasts until one is stably attached to them. Our model reveals new features of how melanoblasts move and, in particular, suggest that melanoblasts leave a repulsive trail behind them as they move through the skin.


Asunto(s)
Movimiento Celular/fisiología , Simulación por Computador , Queratinocitos/metabolismo , Melanocitos/citología , Piel/embriología , Animales , Membrana Basal/metabolismo , Adhesión Celular/fisiología , Matriz Extracelular/metabolismo , Melanocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Teóricos
8.
Genet Med ; 23(9): 1636-1647, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34145395

RESUMEN

PURPOSE: Much of the heredity of melanoma remains unexplained. We sought predisposing germline copy-number variants using a rare disease approach. METHODS: Whole-genome copy-number findings in patients with melanoma predisposition syndrome congenital melanocytic nevus were extrapolated to a sporadic melanoma cohort. Functional effects of duplications in PPP2R3B were investigated using immunohistochemistry, transcriptomics, and stable inducible cellular models, themselves characterized using RNAseq, quantitative real-time polymerase chain reaction (qRT-PCR), reverse phase protein arrays, immunoblotting, RNA interference, immunocytochemistry, proliferation, and migration assays. RESULTS: We identify here a previously unreported genetic susceptibility to melanoma and melanocytic nevi, familial duplications of gene PPP2R3B. This encodes PR70, a regulatory unit of critical phosphatase PP2A. Duplications increase expression of PR70 in human nevus, and increased expression in melanoma tissue correlates with survival via a nonimmunological mechanism. PPP2R3B overexpression induces pigment cell switching toward proliferation and away from migration. Importantly, this is independent of the known microphthalmia-associated transcription factor (MITF)-controlled switch, instead driven by C21orf91. Finally, C21orf91 is demonstrated to be downstream of MITF as well as PR70. CONCLUSION: This work confirms the power of a rare disease approach, identifying a previously unreported copy-number change predisposing to melanocytic neoplasia, and discovers C21orf91 as a potentially targetable hub in the control of phenotype switching.


Asunto(s)
Melanoma , Nevo , Neoplasias Cutáneas , Humanos , Inmunohistoquímica , Melanoma/genética , Fenotipo , Neoplasias Cutáneas/genética
9.
Acta Derm Venereol ; 100(11): adv00139, 2020 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-32346747

RESUMEN

Cutaneous melanoma arises from melanocytes following genetic, epigenetic and allogenetic (i.e. other than epi/genetic) modifications. An estimated 10% of cutaneous melanoma cases are due to inherited variants or de novo mutations in approximately 20 genes, found using linkage, next-generation sequencing and association studies. Based on these studies, 3 classes of predisposing melanoma genes have been defined based on the frequency of the variants in the general population and lifetime risk of developing a melanoma: (i) ultra-rare variants with a high risk, (ii) rare with a moderate risk, and (iii) frequent variants with a low risk. Most of the proteins encoded by these genes have been shown to be involved in melanoma initiation, including proliferation and senescence bypass. This paper reviews the role(s) of these genes in the transformation of melanocytes into melanoma. It also describes their function in the establishment and renewal of melanocytes and the biology of pigment cells, if known.


Asunto(s)
Biomarcadores de Tumor/genética , Melanocitos/patología , Melanoma/genética , Mutación , Neoplasias Cutáneas/genética , Animales , Linaje de la Célula , Predisposición Genética a la Enfermedad , Humanos , Melaninas/metabolismo , Melanocitos/metabolismo , Melanoma/etnología , Melanoma/metabolismo , Melanoma/patología , Melanosomas/metabolismo , Melanosomas/patología , Tasa de Mutación , Fenotipo , Medición de Riesgo , Factores de Riesgo , Neoplasias Cutáneas/etnología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Población Blanca/genética
10.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26240067

RESUMEN

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Asunto(s)
Cadherinas/metabolismo , Transformación Celular Neoplásica , Neoplasias del Colon , Mutación , Proteínas de Neoplasias , Vía de Señalización Wnt , beta Catenina , Animales , Cadherinas/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
11.
Exp Dermatol ; 28(6): 662-666, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30536790

RESUMEN

Vitiligo is a chronic skin disease characterized by the appearance of zones of depigmentation. It is mostly described as an autoimmune disease in which the immune system destroys the melanocytes. Consistent with this origin, genetic studies have implicated genes encoding proteins mediating the immune response targeting melanocytes in the aetiology of this disease, together with proteins specific to these cells. However, the destruction of melanocytes by the immune system is neither global nor complete, because the patients do not display total depigmentation. The etiopathology of vitiligo is clearly complex and cannot be simply reduced to an autoimmune reaction directed against pigmented cells. Intrinsic changes have been observed in the melanocytes, keratinocytes and dermal cells of vitiligo patients. Identification of the molecular and cellular changes occurring in normally pigmented skin in vitiligo patients, and an understanding of these changes, is essential to improve the definition of trigger events for this disease, with a view to developing treatments with long-term efficacy. This review focuses on the early events identified to date in the non-lesional regions of the skin in vitiligo patients and discusses the process of repigmentation from melanocyte stem cells.


Asunto(s)
Melanocitos/inmunología , Vitíligo/inmunología , Apoptosis , Enfermedades Autoinmunes/inmunología , Adhesión Celular , Humanos , Melanocitos/citología , Piel/patología , Células Madre/citología , Células Madre/inmunología
12.
J Math Biol ; 79(6-7): 2111-2132, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515603

RESUMEN

Melanoblast migration is important for embryogenesis and is a key feature of melanoma metastasis. Many studies have characterized melanoblast movement, focusing on statistical properties and have highlighted basic mechanisms of melanoblast motility. We took a slightly different and complementary approach: we previously developed a mathematical model of melanoblast motion that enables the testing of biological assumptions about the displacement of melanoblasts and we created tests to analyze the geometric features of cell trajectories and the specific issue of trajectory interactions. Within this model, we performed simulations and compared the results with experimental data using geometric tests. In this paper, we developed the associated mathematical model and the main focus is to study the crossings between trajectories with new theoretical results about the variation of number of intersection points with respect to the crossing times. Using these results it is possible to study the random nature of displacements and the interactions between trajectories. This analysis has raised new questions, leading to the generation of strong arguments in favor of a trail left behind each moving melanoblast.


Asunto(s)
Movimiento Celular/fisiología , Melanocitos/fisiología , Modelos Biológicos , Células Madre/fisiología , Diferenciación Celular , Desarrollo Embrionario/fisiología , Humanos , Queratinocitos/fisiología , Melanoma/secundario , Neoplasias Cutáneas/patología
13.
J Transl Med ; 16(1): 252, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30285864

RESUMEN

The International Federation of Pigment Cell Societies (IFPCS) held its XXIII triennial International Pigment Cell Conference (IPCC) in Denver, Colorado in August 2017. The goal of the summit was to provide a venue promoting a vibrant interchange among leading basic and clinical researchers working on leading-edge aspects of melanocyte biology and disease. The philosophy of the meeting, entitled Breakthroughs in Pigment Cell and Melanoma Research, was to deliver a comprehensive program in an inclusive environment fostering scientific exchange and building new academic bridges. This document provides an outlook on the history, accomplishments, and sustainability of the pigment cell and melanoma research community. Shared progress in the understanding of cellular homeostasis of pigment cells but also clinical successes and hurdles in the treatment of melanoma and dermatological disorders continue to drive future research activities. A sustainable direction of the societies creates an international forum identifying key areas of imminent needs in laboratory research and clinical care and ensures the future of this vibrant, diverse and unique research community at the same time. Important advances showcase wealth and breadth of the field in melanocyte and melanoma research and include emerging frontiers in melanoma immunotherapy, medical and surgical oncology, dermatology, vitiligo, albinism, genomics and systems biology, precision bench-to-bedside approaches, epidemiology, pigment biophysics and chemistry, and evolution. This report recapitulates highlights of the federate meeting agenda designed to advance clinical and basic research frontiers from melanoma and dermatological sciences followed by a historical perspective of the associated societies and conferences.


Asunto(s)
Internacionalidad , Melanocitos/patología , Distinciones y Premios , Humanos
14.
PLoS Genet ; 11(10): e1005555, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26440048

RESUMEN

MIcrophthalmia-associated Transcription Factor (MITF) regulates melanocyte and melanoma physiology. We show that MITF associates the NURF chromatin-remodelling factor in melanoma cells. ShRNA-mediated silencing of the NURF subunit BPTF revealed its essential role in several melanoma cell lines and in untransformed melanocytes in vitro. Comparative RNA-seq shows that MITF and BPTF co-regulate overlapping gene expression programs in cell lines in vitro. Somatic and specific inactivation of Bptf in developing murine melanoblasts in vivo shows that Bptf regulates their proliferation, migration and morphology. Once born, Bptf-mutant mice display premature greying where the second post-natal coat is white. This second coat is normally pigmented by differentiated melanocytes derived from the adult melanocyte stem cell (MSC) population that is stimulated to proliferate and differentiate at anagen. An MSC population is established and maintained throughout the life of the Bptf-mutant mice, but these MSCs are abnormal and at anagen, give rise to reduced numbers of transient amplifying cells (TACs) that do not express melanocyte markers and fail to differentiate into mature melanin producing melanocytes. MSCs display a transcriptionally repressed chromatin state and Bptf is essential for reactivation of the melanocyte gene expression program at anagen, the subsequent normal proliferation of TACs and their differentiation into mature melanocytes.


Asunto(s)
Antígenos Nucleares/genética , Ensamble y Desensamble de Cromatina/genética , Melanoma/genética , Células Madre Mesenquimatosas , Factor de Transcripción Asociado a Microftalmía/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , División Celular/genética , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso , Melanocitos/metabolismo , Melanoma/patología , Ratones
15.
Int J Cancer ; 141(12): 2551-2561, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28833076

RESUMEN

Targeted oncogene inactivation by small molecule inhibitors can be very effective but tumor recurrence is a frequent problem in the clinic. Therapy by inactivation of the cancer-driving oncogene in transplanted tumors was shown to be augmented in the presence of T cells. However, these experiments did not take into account the long-term, usually tolerogenic, interaction of de novo malignancies with the immune system. Here, we employed mice, in which SV40 large T (Tag) and firefly luciferase (Luc) as fusion protein (TagLuc) could be regulated with the Tet-on system and upon activation resulted in tumors after a long latency. TagLuc inactivation induced profound tumor regression, demonstrating sustained oncogene addiction. While tumor relapse after TagLuc inactivation was prevented in immunocompetent mice bearing transplanted tumors, autochthonous tumors relapsed or recurred after therapy discontinuation indicating that the immune system that coevolved with the malignancy over an extended period of time lost the potency to mount an efficient anti-tumor immune response. By contrast, adoptively transferred CD8+ T cells targeting the cancer-driving oncogene eradicated recurrent autochthonous tumors, highlighting a suitable therapy option in a clinically relevant model.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Doxiciclina/administración & dosificación , Silenciador del Gen , Sistema Inmunológico/metabolismo , Neoplasias Experimentales/terapia , Traslado Adoptivo , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Doxiciclina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Proteínas Recombinantes de Fusión/genética
16.
J Cell Sci ; 128(15): 2938-50, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26092939

RESUMEN

The v-ATPase is a fundamental eukaryotic enzyme that is central to cellular homeostasis. Although its impact on key metabolic regulators such as TORC1 is well documented, our knowledge of mechanisms that regulate v-ATPase activity is limited. Here, we report that the Drosophila transcription factor Mitf is a master regulator of this holoenzyme. Mitf directly controls transcription of all 15 v-ATPase components through M-box cis-sites and this coordinated regulation affects holoenzyme activity in vivo. In addition, through the v-ATPase, Mitf promotes the activity of TORC1, which in turn negatively regulates Mitf. We provide evidence that Mitf, v-ATPase and TORC1 form a negative regulatory loop that maintains each of these important metabolic regulators in relative balance. Interestingly, direct regulation of v-ATPase genes by human MITF also occurs in cells of the melanocytic lineage, showing mechanistic conservation in the regulation of the v-ATPase by MITF family proteins in fly and mammals. Collectively, this evidence points to an ancient module comprising Mitf, v-ATPase and TORC1 that serves as a dynamic modulator of metabolism for cellular homeostasis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Factores de Transcripción/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Drosophila , Activación Enzimática , Homeostasis/fisiología , Humanos , Melanocitos/metabolismo , Melanoma/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño , Transcripción Genética/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo
17.
Exp Dermatol ; 26(10): 875-882, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28191677

RESUMEN

The exposure of skin to ultraviolet (UV) radiation can have both beneficial and deleterious effects: it can lead, for instance, to increased pigmentation and vitamin D synthesis but also to inflammation and skin cancer. UVB may induce genetic and epigenetic alterations and have reversible effects associated with post-translational and gene regulation modifications. ß-catenin is a main driver in melanocyte development; although infrequently mutated in melanoma, its cellular localization and activity are frequently altered. Here, we evaluate the consequence of UVB on ß-catenin in the melanocyte lineage. We report that in vivo, UVB induces cytoplasmic/nuclear relocalization of ß-catenin in melanocytes of newborn mice and adult human skin. In mouse melanocyte and human melanoma cell lines in vitro, UVB increases ß-catenin stability, accumulation in the nucleus and cotranscriptional activity, leading to the repression of cell motility and velocity. The activation of the ß-catenin signalling pathway and its effect on migration by UVB are increased by an inhibitor of GSK3ß, and decreased by an inhibitor of ß-catenin. In conclusion, UVB represses melanocyte migration and does so by acting through the GSK3-ß-catenin axis.


Asunto(s)
Movimiento Celular/efectos de la radiación , Melanocitos/efectos de la radiación , Melanoma/metabolismo , Transporte de Proteínas/efectos de la radiación , Rayos Ultravioleta , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Queratinocitos , Melanocitos/fisiología , Ratones , Fosforilación/efectos de la radiación , Transducción de Señal/efectos de la radiación , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
18.
PLoS Genet ; 10(5): e1004321, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24810760

RESUMEN

Understanding the molecular mechanisms of ultraviolet (UV) induced melanoma formation is becoming crucial with more reported cases each year. Expression of type II nuclear receptor Retinoid-X-Receptor α (RXRα) is lost during melanoma progression in humans. Here, we observed that in mice with melanocyte-specific ablation of RXRα and RXRß, melanocytes attract fewer IFN-γ secreting immune cells than in wild-type mice following acute UVR exposure, via altered expression of several chemoattractive and chemorepulsive chemokines/cytokines. Reduced IFN-γ in the microenvironment alters UVR-induced apoptosis, and due to this, the survival of surrounding dermal fibroblasts is significantly decreased in mice lacking RXRα/ß. Interestingly, post-UVR survival of the melanocytes themselves is enhanced in the absence of RXRα/ß. Loss of RXRs α/ß specifically in the melanocytes results in an endogenous shift in homeostasis of pro- and anti-apoptotic genes in these cells and enhances their survival compared to the wild type melanocytes. Therefore, RXRs modulate post-UVR survival of dermal fibroblasts in a "non-cell autonomous" manner, underscoring their role in immune surveillance, while independently mediating post-UVR melanocyte survival in a "cell autonomous" manner. Our results emphasize a novel immunomodulatory role of melanocytes in controlling survival of neighboring cell types besides controlling their own, and identifies RXRs as potential targets for therapy against UV induced melanoma.


Asunto(s)
Ciclo Celular/efectos de la radiación , Inmunidad Innata/fisiología , Melanocitos/fisiología , Receptor alfa X Retinoide/fisiología , Receptor beta X Retinoide/fisiología , Rayos Ultravioleta , Animales , Melanocitos/efectos de la radiación , Ratones , Ratones Transgénicos , Receptor alfa X Retinoide/genética , Receptor beta X Retinoide/genética
19.
Exp Dermatol ; 25(9): 669-73, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27119971

RESUMEN

Melanocytes arise from the fourth embryonic layer, the neural crest. They emerge from the roof plate of the neural tube and migrate throughout the body. In mammals, these cells have the capacity to migrate in any type of environment and use various pathways and mechanisms to colonize the skin and hair, and for their maintenance throughout the life of the animal.


Asunto(s)
Movimiento Celular , Melanocitos , Piel/embriología , Animales , Humanos
20.
PLoS Genet ; 8(5): e1002688, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22570637

RESUMEN

Studies of coat color mutants have greatly contributed to the discovery of genes that regulate melanocyte development and function. Here, we generated Yy1 conditional knockout mice in the melanocyte-lineage and observed profound melanocyte deficiency and premature gray hair, similar to the loss of melanocytes in human piebaldism and Waardenburg syndrome. Although YY1 is a ubiquitous transcription factor, YY1 interacts with M-MITF, the Waardenburg Syndrome IIA gene and a master transcriptional regulator of melanocytes. YY1 cooperates with M-MITF in regulating the expression of piebaldism gene KIT and multiple additional pigmentation genes. Moreover, ChIP-seq identified genome-wide YY1 targets in the melanocyte lineage. These studies mechanistically link genes implicated in human conditions of melanocyte deficiency and reveal how a ubiquitous factor (YY1) gains lineage-specific functions by co-regulating gene expression with a lineage-restricted factor (M-MITF)-a general mechanism which may confer tissue-specific gene expression in multiple lineages.


Asunto(s)
Color del Cabello , Melanocitos , Factor de Transcripción Asociado a Microftalmía/metabolismo , Pigmentación , Síndrome de Waardenburg , Factor de Transcripción YY1/genética , Animales , Linaje de la Célula , Supervivencia Celular , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Color del Cabello/genética , Humanos , Melanocitos/citología , Melanocitos/metabolismo , Ratones , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/genética , Pigmentación/genética , Síndrome de Waardenburg/genética , Síndrome de Waardenburg/metabolismo , Factor de Transcripción YY1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA