Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(1): 219-235.e21, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31522890

RESUMEN

Although clonal neo-antigen burden is associated with improved response to immune therapy, the functional basis for this remains unclear. Here we study this question in a novel controlled mouse melanoma model that enables us to explore the effects of intra-tumor heterogeneity (ITH) on tumor aggressiveness and immunity independent of tumor mutational burden. Induction of UVB-derived mutations yields highly aggressive tumors with decreased anti-tumor activity. However, single-cell-derived tumors with reduced ITH are swiftly rejected. Their rejection is accompanied by increased T cell reactivity and a less suppressive microenvironment. Using phylogenetic analyses and mixing experiments of single-cell clones, we dissect two characteristics of ITH: the number of clones forming the tumor and their clonal diversity. Our analysis of melanoma patient tumor data recapitulates our results in terms of overall survival and response to immune checkpoint therapy. These findings highlight the importance of clonal mutations in robust immune surveillance and the need to quantify patient ITH to determine the response to checkpoint blockade.


Asunto(s)
Heterogeneidad Genética/efectos de la radiación , Melanoma/genética , Melanoma/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Rayos Ultravioleta/efectos adversos , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Humanos , Linfocitos Infiltrantes de Tumor , Melanoma/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Mutación/efectos de la radiación , Filogenia , Neoplasias Cutáneas/mortalidad , Tasa de Supervivencia , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de la radiación
2.
Cell ; 163(1): 39-53, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26406370

RESUMEN

Significant advances have been made in developing novel therapeutics for cancer treatment, and targeted therapies have revolutionized the treatment of some cancers. Despite the promise, only about five percent of new cancer drugs are approved, and most fail due to lack of efficacy. The indication is that current preclinical methods are limited in predicting successful outcomes. Such failure exacts enormous cost, both financial and in the quality of human life. This Primer explores the current status, promise, and challenges of preclinical evaluation in advanced mouse cancer models and briefly addresses emerging models for early-stage preclinical development.


Asunto(s)
Modelos Animales de Enfermedad , Ratones , Neoplasias/tratamiento farmacológico , Animales , Ingeniería Genética , Xenoinjertos , Trasplante de Neoplasias
3.
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
4.
Am J Hum Genet ; 108(9): 1611-1630, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34343493

RESUMEN

Genome-wide association studies (GWASs) have identified a melanoma-associated locus on chromosome band 7p21.1 with rs117132860 as the lead SNP and a secondary independent signal marked by rs73069846. rs117132860 is also associated with tanning ability and cutaneous squamous cell carcinoma (cSCC). Because ultraviolet radiation (UVR) is a key environmental exposure for all three traits, we investigated the mechanisms by which this locus contributes to melanoma risk, focusing on cellular response to UVR. Fine-mapping of melanoma GWASs identified four independent sets of candidate causal variants. A GWAS region-focused Capture-C study of primary melanocytes identified physical interactions between two causal sets and the promoter of the aryl hydrocarbon receptor (AHR). Subsequent chromatin state annotation, eQTL, and luciferase assays identified rs117132860 as a functional variant and reinforced AHR as a likely causal gene. Because AHR plays critical roles in cellular response to dioxin and UVR, we explored links between this SNP and AHR expression after both 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and ultraviolet B (UVB) exposure. Allele-specific AHR binding to rs117132860-G was enhanced following both, consistent with predicted weakened AHR binding to the risk/poor-tanning rs117132860-A allele, and allele-preferential AHR expression driven from the protective rs117132860-G allele was observed following UVB exposure. Small deletions surrounding rs117132860 introduced via CRISPR abrogates AHR binding, reduces melanocyte cell growth, and prolongs growth arrest following UVB exposure. These data suggest AHR is a melanoma susceptibility gene at the 7p21.1 risk locus and rs117132860 is a functional variant within a UVB-responsive element, leading to allelic AHR expression and altering melanocyte growth phenotypes upon exposure.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma de Células Escamosas/genética , Cromosomas Humanos Par 7 , Sitios Genéticos , Melanocitos/metabolismo , Melanoma/genética , Receptores de Hidrocarburo de Aril/genética , Neoplasias Cutáneas/genética , Alelos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Cromatina/química , Cromatina/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Melanocitos/efectos de los fármacos , Melanocitos/patología , Melanocitos/efectos de la radiación , Melanoma/metabolismo , Melanoma/patología , Dibenzodioxinas Policloradas/toxicidad , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Baño de Sol , Rayos Ultravioleta/efectos adversos
5.
Int J Mol Sci ; 25(9)2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38732242

RESUMEN

Melanoma is the most severe and fatal form of skin cancer, resulting from multiple gene mutations with high intra-tumor and inter-tumor molecular heterogeneity. Treatment options for patients whose disease has progressed beyond the ability for surgical resection rely on currently accepted standard therapies, notably immune checkpoint inhibitors and targeted therapies. Acquired resistance to these therapies and treatment-associated toxicity necessitate exploring novel strategies, especially those that can be personalized for specific patients and/or populations. Here, we review the current landscape and progress of standard therapies and explore what personalized oncology techniques may entail in the scope of melanoma. Our purpose is to provide an up-to-date summary of the tools at our disposal that work to circumvent the common barriers faced when battling melanoma.


Asunto(s)
Melanoma , Medicina de Precisión , Neoplasias Cutáneas , Humanos , Melanoma/genética , Melanoma/terapia , Melanoma/patología , Medicina de Precisión/métodos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Terapia Molecular Dirigida/métodos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Mutación
6.
Nature ; 548(7669): 537-542, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28783722

RESUMEN

Somatic gene mutations can alter the vulnerability of cancer cells to T-cell-based immunotherapies. Here we perturbed genes in human melanoma cells to mimic loss-of-function mutations involved in resistance to these therapies, by using a genome-scale CRISPR-Cas9 library that consisted of around 123,000 single-guide RNAs, and profiled genes whose loss in tumour cells impaired the effector function of CD8+ T cells. The genes that were most enriched in the screen have key roles in antigen presentation and interferon-γ signalling, and correlate with cytolytic activity in patient tumours from The Cancer Genome Atlas. Among the genes validated using different cancer cell lines and antigens, we identified multiple loss-of-function mutations in APLNR, encoding the apelin receptor, in patient tumours that were refractory to immunotherapy. We show that APLNR interacts with JAK1, modulating interferon-γ responses in tumours, and that its functional loss reduces the efficacy of adoptive cell transfer and checkpoint blockade immunotherapies in mouse models. Our results link the loss of essential genes for the effector function of CD8+ T cells with the resistance or non-responsiveness of cancer to immunotherapies.


Asunto(s)
Genes Esenciales/genética , Inmunoterapia , Neoplasias/genética , Neoplasias/terapia , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/genética , Apelina/metabolismo , Receptores de Apelina/genética , Receptores de Apelina/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Femenino , Genoma/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón gamma/inmunología , Janus Quinasa 1/metabolismo , Bases del Conocimiento , Melanoma/genética , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/terapia , Ratones , Mutación , Neoplasias/inmunología , Neoplasias/metabolismo , Reproducibilidad de los Resultados , Linfocitos T Citotóxicos/metabolismo
7.
Br J Cancer ; 124(1): 161-165, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33024267

RESUMEN

NME1 is a metastasis-suppressor gene (MSG), capable of suppressing metastatic activity in cell lines of melanoma, breast carcinoma and other cancer origins without affecting their growth in culture or as primary tumours. Herein, we selectively ablated the tandemly arranged Nme1 and Nme2 genes to assess their individual impacts on metastatic activity in a mouse model (HGF:p16-/-) of ultraviolet radiation (UVR)-induced melanoma. Metastatic activity was strongly enhanced in both genders of Nme1- and Nme2-null mice, with stronger activity in females across all genotypes. The study ascribes MSG activity to Nme2 for the first time in an in vivo model of spontaneous cancer, as well as a novel metastasis-suppressor function to Nme1 in the specific context of UVR-induced melanoma.


Asunto(s)
Genes Supresores de Tumor , Melanoma/genética , Melanoma/patología , Nucleósido Difosfato Quinasas NM23/genética , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Rayos Ultravioleta/efectos adversos
8.
Bioinformatics ; 36(Suppl_1): i169-i176, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32657358

RESUMEN

MOTIVATION: Recent advances in single-cell sequencing (SCS) offer an unprecedented insight into tumor emergence and evolution. Principled approaches to tumor phylogeny reconstruction via SCS data are typically based on general computational methods for solving an integer linear program, or a constraint satisfaction program, which, although guaranteeing convergence to the most likely solution, are very slow. Others based on Monte Carlo Markov Chain or alternative heuristics not only offer no such guarantee, but also are not faster in practice. As a result, novel methods that can scale up to handle the size and noise characteristics of emerging SCS data are highly desirable to fully utilize this technology. RESULTS: We introduce PhISCS-BnB (phylogeny inference using SCS via branch and bound), a branch and bound algorithm to compute the most likely perfect phylogeny on an input genotype matrix extracted from an SCS dataset. PhISCS-BnB not only offers an optimality guarantee, but is also 10-100 times faster than the best available methods on simulated tumor SCS data. We also applied PhISCS-BnB on a recently published large melanoma dataset derived from the sublineages of a cell line involving 20 clones with 2367 mutations, which returned the optimal tumor phylogeny in <4 h. The resulting phylogeny agrees with and extends the published results by providing a more detailed picture on the clonal evolution of the tumor. AVAILABILITY AND IMPLEMENTATION: https://github.com/algo-cancer/PhISCS-BnB. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Neoplasias , Humanos , Cadenas de Markov , Neoplasias/genética , Filogenia , Análisis de Secuencia , Programas Informáticos
9.
PLoS Biol ; 16(5): e2003648, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29723194

RESUMEN

Melanocyte stem cells (McSCs) and mouse models of hair graying serve as useful systems to uncover mechanisms involved in stem cell self-renewal and the maintenance of regenerating tissues. Interested in assessing genetic variants that influence McSC maintenance, we found previously that heterozygosity for the melanogenesis associated transcription factor, Mitf, exacerbates McSC differentiation and hair graying in mice that are predisposed for this phenotype. Based on transcriptome and molecular analyses of Mitfmi-vga9/+ mice, we report a novel role for MITF in the regulation of systemic innate immune gene expression. We also demonstrate that the viral mimic poly(I:C) is sufficient to expose genetic susceptibility to hair graying. These observations point to a critical suppressor of innate immunity, the consequences of innate immune dysregulation on pigmentation, both of which may have implications in the autoimmune, depigmenting disease, vitiligo.


Asunto(s)
Células Madre Adultas , Color del Cabello/inmunología , Inmunidad Innata , Melanocitos , Factor de Transcripción Asociado a Microftalmía/fisiología , Animales , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Color del Cabello/genética , Interferón Tipo I/metabolismo , Ratones , Ratones Transgénicos , Poli I-C
10.
Int J Cancer ; 142(8): 1652-1663, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29210059

RESUMEN

Aberrant HGF/Met signaling promotes tumor migration, invasion, and metastasis through both autocrine and non-autocrine mechanisms; however, the molecular downstream signaling mechanisms by which HGF/Met induces metastasis are incompletely understood. We here report that Ezrin expression is stimulated by HGF and correlates with activated HGF/Met, indicating that HGF/Met signaling regulates the expression of Ezrin. We show that HGF/Met signaling activates the transcription factor Sp1 through the MAPK pathway, and activated Sp1 can in turn directly bind to the promoter of Ezrin gene and regulate its transcription. Notably, knockdown of Ezrin expression by shRNAs inhibits the metastasis induced by either HGF/Met autocrine or non-autocrine signaling in syngeneic wildtype and HGF transgenic mouse hosts. We also used small molecule drugs in preclinical mouse models to confirm that Ezrin is one of the downstream molecules mediating HGF/Met signaling-induced metastasis in melanoma. We conclude that Ezrin is a key downstream factor involved in the regulation of HGF/Met signaling-induced metastasis and demonstrate a link between Ezrin and HGF/Met/MAPK/Sp1 activation in the metastatic process. Our data indicate that Ezrin represents a promising therapeutic target for patients bearing tumors with activated HGF/Met signaling.


Asunto(s)
Comunicación Autocrina/genética , Proteínas del Citoesqueleto/genética , Factor de Crecimiento de Hepatocito/genética , Melanoma/genética , Melanoma/patología , Metástasis de la Neoplasia/genética , Proteínas Proto-Oncogénicas c-met/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/patología , Regiones Promotoras Genéticas/genética , Transducción de Señal/genética
11.
Nature ; 542(7639): 36-38, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28150762

Asunto(s)
Medicina , Neoplasias , Humanos
12.
PLoS Genet ; 11(1): e1004877, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25629959

RESUMEN

Melanoma is the most fatal skin cancer, but the etiology of this devastating disease is still poorly understood. Recently, the transcription factor Sox10 has been shown to promote both melanoma initiation and progression. Reducing SOX10 expression levels in human melanoma cells and in a genetic melanoma mouse model, efficiently abolishes tumorigenesis by inducing cell cycle exit and apoptosis. Here, we show that this anti-tumorigenic effect functionally involves SOX9, a factor related to SOX10 and upregulated in melanoma cells upon loss of SOX10. Unlike SOX10, SOX9 is not required for normal melanocyte stem cell function, the formation of hyperplastic lesions, and melanoma initiation. To the contrary, SOX9 overexpression results in cell cycle arrest, apoptosis, and a gene expression profile shared by melanoma cells with reduced SOX10 expression. Moreover, SOX9 binds to the SOX10 promoter and induces downregulation of SOX10 expression, revealing a feedback loop reinforcing the SOX10 low/SOX9 high ant,m/ii-tumorigenic program. Finally, SOX9 is required in vitro and in vivo for the anti-tumorigenic effect achieved by reducing SOX10 expression. Thus, SOX10 and SOX9 are functionally antagonistic regulators of melanoma development.


Asunto(s)
Carcinogénesis/genética , Melanoma/genética , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXE/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Folículo Piloso , Humanos , Melanocitos/patología , Melanoma/patología , Ratones , ARN Interferente Pequeño , Factor de Transcripción SOX9/biosíntesis , Factores de Transcripción SOXE/biosíntesis
13.
Int J Mol Sci ; 19(2)2018 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-29385676

RESUMEN

Melanoma remains mostly an untreatable fatal disease despite advances in decoding cancer genomics and developing new therapeutic modalities. Progress in patient care would benefit from additional predictive models germane for human disease mechanisms, tumor heterogeneity, and therapeutic responses. Toward this aim, this review documents comparative aspects of human and naturally occurring canine melanomas. Clinical presentation, pathology, therapies, and genetic alterations are highlighted in the context of current basic and translational research in comparative oncology. Somewhat distinct from sun exposure-related human cutaneous melanomas, there is growing evidence that a variety of gene copy number alterations and protein structure/function mutations play roles in canine melanomas, in circumstances more analogous to human mucosal melanomas and to some extent other melanomas with murine sarcoma viral oncogene homolog B (BRAF), Neuroblastoma RAS Viral (V-Ras) Oncogene Homolog (NRAS), and neurofibromin 1 tumor suppressor NF1 triple wild-type genotype. Gaps in canine genome annotation, as well as an insufficient number and depth of sequences covered, remain considerable barriers to progress and should be collectively addressed. Preclinical approaches can be designed to include canine clinical trials addressing immune modulation as well as combined-targeted inhibition of Rat Sarcoma Superfamily/Mitogen-activated protein kinase (RAS/MAPK) and/or Phosphatidylinositol-3-Kinase/Protein Kinase B/Mammalian target of rapamycin (PI3K/AKT/mTOR) signal transduction, pathways frequently activated in both human and canine melanomas. Future investment should be aimed towards improving understanding of canine melanoma as a predictive preclinical surrogate for human melanoma and for mutually benefiting these uniquely co-dependent species.


Asunto(s)
Enfermedades de los Perros , Sistema de Señalización de MAP Quinasas , Melanoma , Proteínas de Neoplasias , Neoplasias Cutáneas , Animales , Enfermedades de los Perros/genética , Enfermedades de los Perros/inmunología , Enfermedades de los Perros/metabolismo , Enfermedades de los Perros/patología , Perros , Humanos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Melanoma/genética , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Especificidad de la Especie
14.
Lab Invest ; 97(6): 698-705, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28092363

RESUMEN

Melanocytes, a neural crest cell derivative, produce pigment to protect keratinocytes from ultraviolet radiation (UVR). Although melanocytic lesions such as nevi and cutaneous malignant melanomas are known to be associated with sun exposure, the role of UVR in oncogenesis is complex and has yet to be clearly elucidated. UVR appears to have a direct mutational role in inducing or promoting melanoma formation as well as an indirect role through microenvironmental changes. Recent advances in the modeling of human melanoma in animals have built platforms upon which prospective studies can begin to investigate these questions. This review will focus exclusively on genetically engineered mouse models of UVR-induced melanoma. The role that UVR has in mouse models depends on multiple factors, including the waveband, timing, and dose of UVR, as well as the nature of the oncogenic agent(s) driving melanomagenesis in the model. Work in the field has examined the role of neonatal and adult UVR, interactions between UVR and common melanoma oncogenes, the role of sunscreen in preventing melanoma, and the effect of UVR on immune function within the skin. Here we describe relevant mouse models and discuss how these models can best be translated to the study of human skin and cutaneous melanoma.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma , Neoplasias Inducidas por Radiación , Neoplasias Cutáneas , Animales , Humanos , Ratones , Piel/anatomía & histología , Piel/citología , Rayos Ultravioleta
15.
Cancer ; 123(S11): 2089-2103, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28543694

RESUMEN

Melanoma is a complex disease that exhibits highly heterogeneous etiological, histopathological, and genetic features, as well as therapeutic responses. Genetically engineered mouse (GEM) models provide powerful tools to unravel the molecular mechanisms critical for melanoma development and drug resistance. Here, we expound briefly the basis of the mouse modeling design, the available technology for genetic engineering, and the aspects influencing the use of GEMs to model melanoma. Furthermore, we describe in detail the currently available GEM models of melanoma. Cancer 2017;123:2089-103. © 2017 American Cancer Society.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma/genética , Ratones , Neoplasias Cutáneas/genética , Animales , Ingeniería Genética , Ratones Noqueados , Ratones Transgénicos , Transcriptoma
16.
Blood ; 125(5): 793-802, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25499762

RESUMEN

Dyskeratosis congenita (DC) is an inherited multisystem disorder, characterized by oral leukoplakia, nail dystrophy, and abnormal skin pigmentation, as well as high rates of bone marrow (BM) failure, solid tumors, and other medical problems such as osteopenia. DC and telomere biology disorders (collectively referred to as TBD here) are caused by germline mutations in telomere biology genes leading to very short telomeres and limited proliferative potential of hematopoietic stem cells. We found that skeletal stem cells (SSCs) within the BM stromal cell population (BMSCs, also known as BM-derived mesenchymal stem cells), may contribute to the hematologic phenotype. TBD-BMSCs exhibited reduced clonogenicity, spontaneous differentiation into adipocytes and fibrotic cells, and increased senescence in vitro. Upon in vivo transplantation into mice, TBD-BMSCs failed to form bone or support hematopoiesis, unlike normal BMSCs. TERC reduction (a TBD-associated gene) in normal BMSCs by small interfering TERC-RNA (siTERC-RNA) recapitulated the TBD-BMSC phenotype by reducing proliferation and secondary colony-forming efficiency, and by accelerating senescence in vitro. Microarray profiles of control and siTERC-BMSCs showed decreased hematopoietic factors at the messenger RNA level and decreased secretion of factors at the protein level. These findings are consistent with defects in SSCs/BMSCs contributing to BM failure in TBD.


Asunto(s)
Células de la Médula Ósea/metabolismo , Disqueratosis Congénita/genética , Células Madre Mesenquimatosas/metabolismo , ARN/genética , Telomerasa/genética , Telómero/metabolismo , Adolescente , Adulto , Animales , Secuencia de Bases , Células de la Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Senescencia Celular , Niño , Preescolar , Ensayo de Unidades Formadoras de Colonias , ADN Helicasas/genética , ADN Helicasas/metabolismo , Disqueratosis Congénita/patología , Femenino , Hematopoyesis/genética , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , ARN/antagonistas & inhibidores , ARN/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Telomerasa/antagonistas & inhibidores , Telomerasa/metabolismo , Telómero/química , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
17.
Nature ; 469(7331): 548-53, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21248750

RESUMEN

Cutaneous malignant melanoma is a highly aggressive and frequently chemoresistant cancer, the incidence of which continues to rise. Epidemiological studies show that the major aetiological melanoma risk factor is ultraviolet (UV) solar radiation, with the highest risk associated with intermittent burning doses, especially during childhood. We have experimentally validated these epidemiological findings using the hepatocyte growth factor/scatter factor transgenic mouse model, which develops lesions in stages highly reminiscent of human melanoma with respect to biological, genetic and aetiological criteria, but only when irradiated as neonatal pups with UVB, not UVA. However, the mechanisms underlying UVB-initiated, neonatal-specific melanomagenesis remain largely unknown. Here we introduce a mouse model permitting fluorescence-aided melanocyte imaging and isolation following in vivo UV irradiation. We use expression profiling to show that activated neonatal skin melanocytes isolated following a melanomagenic UVB dose bear a distinct, persistent interferon response signature, including genes associated with immunoevasion. UVB-induced melanocyte activation, characterized by aberrant growth and migration, was abolished by antibody-mediated systemic blockade of interferon-γ (IFN-γ), but not type-I interferons. IFN-γ was produced by macrophages recruited to neonatal skin by UVB-induced ligands to the chemokine receptor Ccr2. Admixed recruited skin macrophages enhanced transplanted melanoma growth by inhibiting apoptosis; notably, IFN-γ blockade abolished macrophage-enhanced melanoma growth and survival. IFN-γ-producing macrophages were also identified in 70% of human melanomas examined. Our data reveal an unanticipated role for IFN-γ in promoting melanocytic cell survival/immunoevasion, identifying a novel candidate therapeutic target for a subset of melanoma patients.


Asunto(s)
Interferón gamma/metabolismo , Melanocitos/metabolismo , Melanoma/fisiopatología , Rayos Ultravioleta , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Humanos , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Masculino , Melanocitos/efectos de la radiación , Ratones
18.
Int J Mol Sci ; 18(8)2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28809777

RESUMEN

Pigmented epithelioid melanocytoma (PEM) is a tumor encompassing epithelioid blue nevus of Carney complex (EBN of CNC) and was previously termed animal-type melanoma. Histologically PEMs are heavily pigmented spindled and epithelioid dermal melanocytic tumors with infiltrative borders, however, their origin remains unclear. Stem cells for the epidermis and hair follicle are located in the bulge area of the hair follicle with the potential to differentiate into multiple lineages. Multiple cutaneous carcinomas, including follicular cutaneous squamous cell carcinoma (FSCC), are thought to arise from stem cells in the follicular bulge. We present two cases of PEM/ATM in a 63 year-old male on the scalp with follicular origin and a 72 year-old female on the upper back arising in an intradermal nevus. Biopsy of both cases revealed a proliferation of heavily pigmented dermal nests of melanocytes with atypia. The Case 1 tumor was in continuation with the outer root sheath of the hair follicle in the bulge region. Case 2 arose in an intradermal melanocytic nevus. Rare mitotic figures, including atypical mitotic figures, were identified in both cases. We present two cases of PEM, with histologic evidence suggesting two origins: one from the follicular bulb and one from an intradermal nevus.


Asunto(s)
Neoplasias de Cabeza y Cuello , Melanoma , Nevo Intradérmico , Neoplasias Cutáneas , Anciano , Animales , Folículo Piloso/metabolismo , Folículo Piloso/patología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patología , Nevo Intradérmico/metabolismo , Nevo Intradérmico/patología , Cuero Cabelludo/metabolismo , Cuero Cabelludo/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
19.
Stem Cells ; 33(4): 1304-19, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25524638

RESUMEN

The tumor suppressor, p53, plays a critical role in suppressing osteosarcoma. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested to give rise to osteosarcomas. However, the role of p53 in BMSCs has not been extensively explored. Here, we report that p53 regulates the lineage choice of mouse BMSCs (mBMSCs). Compared to mBMSCs with wild-type p53, mBMSCs deficient in p53 have enhanced osteogenic differentiation, but with similar adipogenic and chondrogenic differentiation. The role of p53 in inhibiting osteogenic lineage differentiation is mainly through the action of Runx2, a master transcription factor required for the osteogenic differentiation of mBMSCs. We find that p53 indirectly represses the expression of Runx2 by activating the microRNA-34 family, which suppresses the translation of Runx2. Since osteosarcoma may derive from BMSCs, we examined whether p53 has a role in the osteogenic differentiation of osteosarcoma cells and found that osteosarcoma cells with p53 deletion have higher levels of Runx2 and faster osteogenic differentiation than those with wild-type p53. A systems biology approach reveals that p53-deficient mBMSCs are more closely related to human osteosarcoma while mBMSCs with wild-type p53 are similar to normal human BMSCs. In summary, our results indicate that p53 activity can influence cell fate specification of mBMSCs, and provide molecular and cellular insights into the observation that p53 loss is associated with increased osteosarcoma incidence.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Animales , Línea Celular Tumoral , Células Cultivadas , Humanos , Ratones , Ratones Noqueados
20.
Int J Cancer ; 136(4): 752-60, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24975581

RESUMEN

Melanocortin 1 receptor (MC1R) signaling stimulates black eumelanin production through a cAMP-dependent pathway. MC1R polymorphisms can impair this process, resulting in a predominance of red phaeomelanin. The red hair, fair skin and UV sensitive phenotype is a well-described melanoma risk factor. MC1R polymorphisms also confer melanoma risk independent of pigment. We investigated the effect of Mc1r deficiency in a mouse model of UV-induced melanoma. C57BL/6-Mc1r+/+-HGF transgenic mice have a characteristic hyperpigmented black phenotype with extra-follicular dermal melanocytes located at the dermal/epidermal junction. UVB induces melanoma, independent of melanin pigmentation, but UVA-induced and spontaneous melanomas are dependent on black eumelanin. We crossed these mice with yellow C57BL/6-Mc1re/e animals which have a non-functional Mc1r and produce predominantly yellow phaeomelanin. Yellow C57BL/6-Mc1re/e-HGF mice produced no melanoma in response to UVR or spontaneously even though the HGF transgene and its receptor Met were expressed. Total melanin was less than in C57BL/6-Mc1r+/+-HGF mice, hyperpigmentation was not observed and there were few extra-follicular melanocytes. Thus, functional Mc1r was required for expression of the transgenic HGF phenotype. Heterozygous C57BL/6-Mc1re/+-HGF mice were black and hyperpigmented and, although extra-follicular melanocytes and skin melanin content were similar to C57BL/6-Mc1r+/+-HGF animals, they developed UV-induced and spontaneous melanomas with significantly less efficiency by all criteria. Thus, heterozygosity for Mc1r was sufficient to restore the transgenic HGF phenotype but insufficient to fully restore melanoma. We conclude that a previously unsuspected melanin-independent interaction between Mc1r and Met signaling pathways is required for HGF-dependent melanoma and postulate that this pathway is involved in human melanoma.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Melanoma Experimental/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Femenino , Humanos , Masculino , Melaninas/fisiología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA