Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Pharmacol Ther ; 248: 108466, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37301330

RESUMO

Melanoma, the cancer of the melanocyte, is the deadliest form of skin cancer with an aggressive nature, propensity to metastasize and tendency to resist therapeutic intervention. Studies have identified that the re-emergence of developmental pathways in melanoma contributes to melanoma onset, plasticity, and therapeutic response. Notably, it is well known that noncoding RNAs play a critical role in the development and stress response of tissues. In this review, we focus on the noncoding RNAs, including microRNAs, long non-coding RNAs, circular RNAs, and other small RNAs, for their functions in developmental mechanisms and plasticity, which drive onset, progression, therapeutic response and resistance in melanoma. Going forward, elucidation of noncoding RNA-mediated mechanisms may provide insights that accelerate development of novel melanoma therapies.


Assuntos
Melanoma , MicroRNAs , RNA Longo não Codificante , Humanos , RNA não Traduzido/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , RNA Longo não Codificante/genética , RNA Circular
2.
bioRxiv ; 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-37333132

RESUMO

Intratumoral heterogeneity (ITH) can promote cancer progression and treatment failure, but the complexity of the regulatory programs and contextual factors involved complicates its study. To understand the specific contribution of ITH to immune checkpoint blockade (ICB) response, we generated single cell-derived clonal sublines from an ICB-sensitive and genetically and phenotypically heterogeneous mouse melanoma model, M4. Genomic and single cell transcriptomic analyses uncovered the diversity of the sublines and evidenced their plasticity. Moreover, a wide range of tumor growth kinetics were observed in vivo , in part associated with mutational profiles and dependent on T cell-response. Further inquiry into melanoma differentiation states and tumor microenvironment (TME) subtypes of untreated tumors from the clonal sublines demonstrated correlations between highly inflamed and differentiated phenotypes with the response to anti-CTLA-4 treatment. Our results demonstrate that M4 sublines generate intratumoral heterogeneity at both levels of intrinsic differentiation status and extrinsic TME profiles, thereby impacting tumor evolution during therapeutic treatment. These clonal sublines proved to be a valuable resource to study the complex determinants of response to ICB, and specifically the role of melanoma plasticity in immune evasion mechanisms.

3.
Dev Cell ; 57(21): 2447-2449, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36347238

RESUMO

Melanoma evolution may recapitulate the embryonic development of its progenitor tissue, neural crest (NC), but the exact process is unclear. In a recent issue of Nature, Karras et al. (2022) demonstrate that melanoma expansion mirrors the hierarchic process of NC differentiation, generating cell subpopulations, each with distinct function, including growth and metastasis.


Assuntos
Melanoma , Humanos , Diferenciação Celular , Crista Neural , Organogênese
4.
Nat Comput Sci ; 2(9): 577-583, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38177468

RESUMO

We introduce HUNTRESS, a computational method for mutational intratumor heterogeneity inference from noisy genotype matrices derived from single-cell sequencing data, the running time of which is linear with the number of cells and quadratic with the number of mutations. We prove that, under reasonable conditions, HUNTRESS computes the true progression history of a tumor with high probability. On simulated and real tumor sequencing data, HUNTRESS is demonstrated to be faster than available alternatives with comparable or better accuracy. Additionally, the progression histories of tumors inferred by HUNTRESS on real single-cell sequencing datasets agree with the best known evolution scenarios for the associated tumors.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Análise de Sequência , Mutação
5.
Cancer Cell ; 39(5): 610-631, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33545064

RESUMO

There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.


Assuntos
Modelos Animais de Doenças , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Microambiente Tumoral/imunologia , Animais , Humanos , Imunidade/imunologia , Imunoterapia/métodos , Melanoma/patologia , Neoplasias Cutâneas/patologia
6.
Dev Cell ; 54(3): 317-332.e9, 2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32652076

RESUMO

Melanocytes, replenished throughout life by melanocyte stem cells (MSCs), play a critical role in pigmentation and melanoma. Here, we reveal a function for the metastasis-associated phosphatase of regenerating liver 3 (PRL3) in MSC regeneration. We show that PRL3 binds to the RNA helicase DDX21, thereby restricting productive transcription by RNAPII at master transcription factor (MITF)-regulated endolysosomal vesicle genes. In zebrafish, this mechanism controls premature melanoblast expansion and differentiation from MSCs. In melanoma patients, restricted transcription of this endolysosomal vesicle pathway is a hallmark of PRL3-high melanomas. Our work presents the conceptual advance that PRL3-mediated control of transcriptional elongation is a differentiation checkpoint mechanism for activated MSCs and has clinical relevance for the activity of PRL3 in regenerating tissue and cancer.


Assuntos
Diferenciação Celular/genética , RNA Helicases DEAD-box/metabolismo , Melanócitos/citologia , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , Mutação , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatases/genética , Células-Tronco/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
7.
Nat Commun ; 11(1): 333, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31949145

RESUMO

Cutaneous malignant melanoma is an aggressive cancer of melanocytes with a strong propensity to metastasize. We posit that melanoma cells acquire metastatic capability by adopting an embryonic-like phenotype, and that a lineage approach would uncover metastatic melanoma biology. Using a genetically engineered mouse model to generate a rich melanoblast transcriptome dataset, we identify melanoblast-specific genes whose expression contribute to metastatic competence and derive a 43-gene signature that predicts patient survival. We identify a melanoblast gene, KDELR3, whose loss impairs experimental metastasis. In contrast, KDELR1 deficiency enhances metastasis, providing the first example of different disease etiologies within the KDELR-family of retrograde transporters. We show that KDELR3 regulates the metastasis suppressor, KAI1, and report an interaction with the E3 ubiquitin-protein ligase gp78, a regulator of KAI1 degradation. Our work demonstrates that the melanoblast transcriptome can be mined to uncover targetable pathways for melanoma therapy.


Assuntos
Perfilação da Expressão Gênica , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transcriptoma , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Pulmão/patologia , Melanócitos/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/genética , Segunda Neoplasia Primária/patologia , Fenótipo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Neoplasias Cutâneas/patologia , Ubiquitina-Proteína Ligases/metabolismo , Melanoma Maligno Cutâneo
8.
Nat Med ; 26(5): 781-791, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32284588

RESUMO

Although immunotherapy has revolutionized cancer treatment, only a subset of patients demonstrate durable clinical benefit. Definitive predictive biomarkers and targets to overcome resistance remain unidentified, underscoring the urgency to develop reliable immunocompetent models for mechanistic assessment. Here we characterize a panel of syngeneic mouse models, representing a variety of molecular and phenotypic subtypes of human melanomas and exhibiting their diverse range of responses to immune checkpoint blockade (ICB). Comparative analysis of genomic, transcriptomic and tumor-infiltrating immune cell profiles demonstrated alignment with clinical observations and validated the correlation of T cell dysfunction and exclusion programs with resistance. Notably, genome-wide expression analysis uncovered a melanocytic plasticity signature predictive of patient outcome in response to ICB, suggesting that the multipotency and differentiation status of melanoma can determine ICB benefit. Our comparative preclinical platform recapitulates melanoma clinical behavior and can be employed to identify mechanisms and treatment strategies to improve patient care.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais , Imunoterapia , Melanoma/patologia , Melanoma/terapia , Animais , Antineoplásicos Imunológicos/uso terapêutico , Antígeno CTLA-4/imunologia , Células Cultivadas , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Heterogeneidade Genética , Humanos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Ipilimumab/uso terapêutico , Melanoma/diagnóstico , Melanoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Prognóstico , Receptor de Morte Celular Programada 1/imunologia , RNA-Seq , Resultado do Tratamento , Sequenciamento Completo do Genoma
9.
Cancer Discov ; 9(1): 19-21, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30626604

RESUMO

In this issue, Weeraratna and colleagues demonstrate that observed differences in melanoma aggressiveness in younger versus older patients can be explained not just by cell-intrinsic alterations over time, but by age-dependent changes in fibroblasts and the extracellular matrix they help create. Their findings identify novel cellular targets for melanoma therapy, as well as candidate prognostic biomarkers to better inform clinical decisions for patients with melanoma.See related article by Kaur et al., p. 64.See related article by Ecker et al., p. 82.


Assuntos
Vasos Linfáticos , Melanoma , Fibroblastos , Humanos , Permeabilidade , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA