Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Trends Cancer ; 10(4): 356-368, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38135572

RESUMO

Epithelial cancers have served as a paradigm to study tumor dissemination but recent data have highlighted significant differences with nonepithelial cancers. Here, we review the current knowledge on nonepithelial tumor dissemination, drawing examples from the latest developments in melanoma, glioma, and sarcoma research. We underscore the importance of the reactivation of developmental processes during cancer progression and describe the nongenetic mechanisms driving nonepithelial tumor spread. We also outline therapeutic opportunities and ongoing clinical approaches to fight disseminating cancers. Finally, we discuss remaining challenges and emerging questions in the field. Defining the core principles underlying nonepithelial cancer dissemination may uncover actionable vulnerabilities of metastatic tumors and help improve the prognosis of patients with cancer.


Assuntos
Glioma , Melanoma , Humanos , Melanoma/terapia , Melanoma/patologia , Microambiente Tumoral
2.
Life Sci Alliance ; 6(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37536978

RESUMO

Triple-negative breast cancers (TNBCs) are the most aggressive breast cancers, and therapeutic options mainly rely on chemotherapy and immunotherapy. Although synthetic glucocorticoids (GCs) are given to alleviate the side effects of these treatments, GCs and their receptor, the glucocorticoid receptor (GR), were recently associated with detrimental effects, albeit the mechanisms involved remain elusive. Here, we identified the arginine methyltransferase PRMT5 as a master coregulator of GR, serving as a scaffold protein to recruit phospho-HP1γ and subsequently RNA polymerase II, independently of its methyltransferase activity. Moreover, the GR/PRMT5/HP1γ complex regulated the transcription of GC-target genes involved in cell motility and triggering cell migration of human TNBC cells in vitro and in a zebrafish model. Of note, we observed that GR/PRMT5 interaction was low in primary tumors but significantly increased in residual tumors treated with chemotherapy and GCs in neoadjuvant setting. These data suggest that the routine premedication prescription of GCs for early TNBC patients should be further assessed and that this complex could potentially be modulated to specifically target deleterious GR effects.


Assuntos
Movimento Celular , Glucocorticoides , Proteína-Arginina N-Metiltransferases , Neoplasias de Mama Triplo Negativas , Movimento Celular/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/patologia , Glucocorticoides/efeitos adversos , Humanos , Animais , Peixe-Zebra , Receptores de Glucocorticoides/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Linhagem Celular Tumoral
3.
Nat Genet ; 54(12): 1839-1852, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36229674

RESUMO

Cancer genetics has uncovered many tumor-suppressor and oncogenic pathways, but few alterations have revealed mechanisms involved in tumor spreading. Here, we examined the role of the third most significant chromosomal deletion in human melanoma that inactivates the adherens junction gene NECTIN1 in 55% of cases. We found that NECTIN1 loss stimulates melanoma cell migration in vitro and spreading in vivo in both zebrafish and human tumors specifically in response to decreased IGF1 signaling. In human melanoma biopsy specimens, adherens junctions were seen exclusively in areas with low IGF1 levels, but not in NECTIN1-deficient tumors. Our study establishes NECTIN1 as a major determinant of melanoma dissemination and uncovers a genetic control of the response to microenvironmental signals.


Assuntos
Melanoma , Peixe-Zebra , Humanos , Animais , Peixe-Zebra/genética , Melanoma/genética , Fator de Crescimento Insulin-Like I/genética
4.
Dis Model Mech ; 15(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35234863

RESUMO

Activating mutations in RAS genes are the most common genetic driver of human cancers. Yet, drugging this small GTPase has proven extremely challenging and therapeutic strategies targeting these recurrent alterations have long had limited success. To circumvent this difficulty, research has focused on the molecular dissection of the RAS pathway to gain a more-precise mechanistic understanding of its regulation, with the hope to identify new pharmacological approaches. Here, we review the current knowledge on the (dys)regulation of the RAS pathway, using melanoma as a paradigm. We first present a map of the main proteins involved in the RAS pathway, highlighting recent insights into their molecular roles and diverse mechanisms of regulation. We then overview genetic data pertaining to RAS pathway alterations in melanoma, along with insight into other cancers, that inform the biological function of members of the pathway. Finally, we describe the clinical implications of RAS pathway dysregulation in melanoma, discuss past and current approaches aimed at drugging the RAS pathway, and outline future opportunities for therapeutic development.


Assuntos
Antineoplásicos , Melanoma , Neoplasias Cutâneas , Proteínas ras , Antineoplásicos/farmacologia , Genes ras , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Mutação/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
5.
Elife ; 102021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33527896

RESUMO

Recent genomic and scRNA-seq analyses of melanoma demonstrated a lack of recurrent genetic drivers of metastasis, while identifying common transcriptional states correlating with invasion or drug resistance. To test whether transcriptional adaptation can drive melanoma progression, we made use of a zebrafish mitfa:BRAFV600E;tp53-/- model, in which malignant progression is characterized by minimal genetic evolution. We undertook an overexpression-screen of 80 epigenetic/transcriptional regulators and found neural crest-mesenchyme developmental regulator SATB2 to accelerate aggressive melanoma development. Its overexpression induces invadopodia formation and invasion in zebrafish tumors and human melanoma cell lines. SATB2 binds and activates neural crest-regulators, including pdgfab and snai2. The transcriptional program induced by SATB2 overlaps with known MITFlowAXLhigh and AQP1+NGFR1high drug-resistant states and functionally drives enhanced tumor propagation and resistance to Vemurafenib in vivo. In summary, we show that melanoma transcriptional rewiring by SATB2 to a neural crest mesenchyme-like program can drive invasion and drug resistance in autochthonous tumors.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Melanoma/genética , Invasividade Neoplásica/genética , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/genética , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Crista Neural/citologia , Fatores de Transcrição/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
6.
J Exp Med ; 218(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33306107

RESUMO

Functional evaluation of genetic lesions can discover a role in cancer initiation and progression and help develop novel therapeutic strategies. We previously identified the negative MAPK regulator SPRED1 as a novel tumor suppressor in KIT-driven melanoma. Here, we show that SPRED1 is also frequently deleted in human melanoma driven by mutant BRAF. We found that SPRED1 inactivation in human melanoma cell lines and primary zebrafish melanoma conferred resistance to BRAFV600E inhibition in vitro and in vivo. Mechanistically, SPRED1 loss promoted melanoma cell proliferation under mutant BRAF inhibition by reactivating MAPK activity. Consistently, biallelic deletion of SPRED1 was observed in a patient whose melanoma acquired resistance to MAPK-targeted therapy. These studies combining work in human cells and in vivo modeling in zebrafish demonstrate a new mechanism of resistance to BRAFV600E inhibition in melanoma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Resistencia a Medicamentos Antineoplásicos/genética , Deleção de Genes , Sistema de Sinalização das MAP Quinases , Melanoma/enzimologia , Melanoma/genética , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra
7.
Nat Commun ; 11(1): 2718, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483191

RESUMO

Genome-wide association studies (GWAS) have identified ~20 melanoma susceptibility loci, most of which are not functionally characterized. Here we report an approach integrating massively-parallel reporter assays (MPRA) with cell-type-specific epigenome and expression quantitative trait loci (eQTL) to identify susceptibility genes/variants from multiple GWAS loci. From 832 high-LD variants, we identify 39 candidate functional variants from 14 loci displaying allelic transcriptional activity, a subset of which corroborates four colocalizing melanocyte cis-eQTL genes. Among these, we further characterize the locus encompassing the HIV-1 restriction gene, MX2 (Chr21q22.3), and validate a functional intronic variant, rs398206. rs398206 mediates the binding of the transcription factor, YY1, to increase MX2 levels, consistent with the cis-eQTL of MX2 in primary human melanocytes. Melanocyte-specific expression of human MX2 in a zebrafish model demonstrates accelerated melanoma formation in a BRAFV600E background. Our integrative approach streamlines GWAS follow-up studies and highlights a pleiotropic function of MX2 in melanoma susceptibility.


Assuntos
Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla/métodos , Melanoma/genética , Mutação , Proteínas de Resistência a Myxovirus/genética , Polimorfismo de Nucleotídeo Único , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genes Reporter/genética , Células HEK293 , Humanos , Melanócitos/metabolismo , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Locos de Características Quantitativas/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
8.
Nat Rev Cancer ; 20(5): 263-273, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32251397

RESUMO

In precision oncology, two major strategies are being pursued for predicting clinically relevant tumour behaviours, such as treatment response and emergence of drug resistance: inference based on genomic, transcriptomic, epigenomic and/or proteomic analysis of patient samples, and phenotypic assays in personalized cancer avatars. The latter approach has historically relied on in vivo mouse xenografts and in vitro organoids or 2D cell cultures. Recent progress in rapid combinatorial genetic modelling, the development of a genetically immunocompromised strain for xenotransplantation of human patient samples in adult zebrafish and the first clinical trial using xenotransplantation in zebrafish larvae for phenotypic testing of drug response bring this tiny vertebrate to the forefront of the precision medicine arena. In this Review, we discuss advances in transgenic and transplantation-based zebrafish cancer avatars, and how these models compare with and complement mouse xenografts and human organoids. We also outline the unique opportunities that these different models present for prediction studies and current challenges they face for future clinical deployment.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Peixe-Zebra , Animais , Modelos Animais de Doenças , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Medicina de Precisão , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
9.
Dev Biol ; 449(2): 107-114, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29883661

RESUMO

NRAS mutations are frequently found in many deadly malignancies and are the second most common oncogene driving malignant melanoma. Here, we generate a rapid transient transgenic zebrafish model of NRASQ61R-mutant melanoma. These fish develop extensive melanocytic proliferation in approximately 4 weeks. The majority of these lesions do not engraft upon transplantation and lack overt histologic features of malignancy. Our previous work demonstrated that activation of a neural crest cell transcriptional program is a key initiating event in zebrafish BRAF/p53-driven melanomas using the fluorescent reporter crestin:EGFP. By 8-12 weeks of age, some lesions progress to malignant melanoma and have cytologic atypia, destructive tissue invasion, and express neural crest progenitor markers, including crestin:EGFP. Our studies demonstrate that NRASQ61R induces extensive melanocyte expansion, which arise during zebrafish development and lack a transformed phenotype. These early lesions are highly predisposed to reactivate a neural crest progenitor fate and form malignant melanomas.


Assuntos
Proliferação de Células/genética , Genes ras/genética , Melanócitos/metabolismo , Melanoma/genética , Mutação , Crista Neural/metabolismo , Neoplasias Cutâneas/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Humanos , Estimativa de Kaplan-Meier , Melanócitos/patologia , Melanoma/metabolismo , Melanoma/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Tempo , Peixe-Zebra , Melanoma Maligno Cutâneo
10.
Science ; 362(6418): 1055-1060, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30385465

RESUMO

Melanomas originating from mucosal surfaces have low mutation burden, genomic instability, and poor prognosis. To identify potential driver genes, we sequenced hundreds of cancer-related genes in 43 human mucosal melanomas, cataloging point mutations, amplifications, and deletions. The SPRED1 gene, which encodes a negative regulator of mitogen-activated protein kinase (MAPK) signaling, was inactivated in 37% of the tumors. Four distinct genotypes were associated with SPRED1 loss. Using a rapid, tissue-specific CRISPR technique to model these genotypes in zebrafish, we found that SPRED1 functions as a tumor suppressor, particularly in the context of KIT mutations. SPRED1 knockdown caused MAPK activation, increased cell proliferation, and conferred resistance to drugs inhibiting KIT tyrosine kinase activity. These findings provide a rationale for MAPK inhibition in SPRED1-deficient melanomas and introduce a zebrafish modeling approach that can be used more generally to dissect genetic interactions in cancer.


Assuntos
Genes Neoplásicos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Melanoma/genética , Proteínas de Membrana/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Cutâneas/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Resistencia a Medicamentos Antineoplásicos/genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genômica , Humanos , Melanoma/patologia , Melanoma Experimental/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Mucosa/enzimologia , Mucosa/patologia , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais , Neoplasias Cutâneas/patologia , Peixe-Zebra
11.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30366982

RESUMO

Erythropoietin (EPO) signaling is critical to many processes essential to terminal erythropoiesis. Despite the centrality of iron metabolism to erythropoiesis, the mechanisms by which EPO regulates iron status are not well-understood. To this end, here we profiled gene expression in EPO-treated 32D pro-B cells and developing fetal liver erythroid cells to identify additional iron regulatory genes. We determined that FAM210B, a mitochondrial inner-membrane protein, is essential for hemoglobinization, proliferation, and enucleation during terminal erythroid maturation. Fam210b deficiency led to defects in mitochondrial iron uptake, heme synthesis, and iron-sulfur cluster formation. These defects were corrected with a lipid-soluble, small-molecule iron transporter, hinokitiol, in Fam210b-deficient murine erythroid cells and zebrafish morphants. Genetic complementation experiments revealed that FAM210B is not a mitochondrial iron transporter but is required for adequate mitochondrial iron import to sustain heme synthesis and iron-sulfur cluster formation during erythroid differentiation. FAM210B was also required for maximal ferrochelatase activity in differentiating erythroid cells. We propose that FAM210B functions as an adaptor protein that facilitates the formation of an oligomeric mitochondrial iron transport complex, required for the increase in iron acquisition for heme synthesis during terminal erythropoiesis. Collectively, our results reveal a critical mechanism by which EPO signaling regulates terminal erythropoiesis and iron metabolism.


Assuntos
Células Eritroides/metabolismo , Eritropoetina/metabolismo , Ferroquelatase/metabolismo , Heme/biossíntese , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Células Eritroides/citologia , Eritropoese , Células HEK293 , Humanos , Proteínas de Membrana/química , Camundongos , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/química , Transporte Proteico
12.
Mol Cell ; 62(1): 34-46, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058786

RESUMO

Studying cancer metabolism gives insight into tumorigenic survival mechanisms and susceptibilities. In melanoma, we identify HEXIM1, a transcription elongation regulator, as a melanoma tumor suppressor that responds to nucleotide stress. HEXIM1 expression is low in melanoma. Its overexpression in a zebrafish melanoma model suppresses cancer formation, while its inactivation accelerates tumor onset in vivo. Knockdown of HEXIM1 rescues zebrafish neural crest defects and human melanoma proliferation defects that arise from nucleotide depletion. Under nucleotide stress, HEXIM1 is induced to form an inhibitory complex with P-TEFb, the kinase that initiates transcription elongation, to inhibit elongation at tumorigenic genes. The resulting alteration in gene expression also causes anti-tumorigenic RNAs to bind to and be stabilized by HEXIM1. HEXIM1 plays an important role in inhibiting cancer cell-specific gene transcription while also facilitating anti-cancer gene expression. Our study reveals an important role for HEXIM1 in coupling nucleotide metabolism with transcriptional regulation in melanoma.


Assuntos
Melanoma/metabolismo , Fator B de Elongação Transcricional Positiva/genética , Pirimidinas/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/patologia , Melanoma Experimental , Proteínas Oncogênicas/genética , Fatores de Transcrição , Transcrição Gênica , Proteínas Supressoras de Tumor/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
Science ; 351(6272): aad2197, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26823433

RESUMO

The "cancerized field" concept posits that cancer-prone cells in a given tissue share an oncogenic mutation, but only discreet clones within the field initiate tumors. Most benign nevi carry oncogenic BRAF(V600E) mutations but rarely become melanoma. The zebrafish crestin gene is expressed embryonically in neural crest progenitors (NCPs) and specifically reexpressed in melanoma. Live imaging of transgenic zebrafish crestin reporters shows that within a cancerized field (BRAF(V600E)-mutant; p53-deficient), a single melanocyte reactivates the NCP state, revealing a fate change at melanoma initiation in this model. NCP transcription factors, including sox10, regulate crestin expression. Forced sox10 overexpression in melanocytes accelerated melanoma formation, which is consistent with activation of NCP genes and super-enhancers leading to melanoma. Our work highlights NCP state reemergence as a key event in melanoma initiation.


Assuntos
Carcinogênese/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/genética , Melanoma/genética , Crista Neural/metabolismo , Neoplasias Cutâneas/genética , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos , Genes Reporter , Proteínas de Fluorescência Verde/genética , Melanócitos/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas B-raf/genética , Fatores de Transcrição SOXE/genética , Proteína Supressora de Tumor p53/genética , Proteínas de Peixe-Zebra/genética
14.
Cold Spring Harb Perspect Med ; 6(1): a026260, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26637438

RESUMO

Although integrity of the p53 signaling pathway in a given tumor was expected to be a critical determinant of response to therapies, most clinical studies failed to link p53 status and treatment outcome. Here, we present two opposite situations: one in which p53 is an essential effector of cure by targeted leukemia therapies and another one in advanced breast cancers in which p53 inactivation is required for the clinical efficacy of dose-dense chemotherapy. If p53 promotes or blocks therapy response, therapies must be tailored on its status in individual tumors.


Assuntos
Neoplasias da Mama/metabolismo , Leucemia Promielocítica Aguda/genética , Proteínas de Fusão Oncogênica/genética , Proteína Supressora de Tumor p53/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Trióxido de Arsênio , Arsenicais/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Terapia de Alvo Molecular , Óxidos/administração & dosagem , Resultado do Tratamento , Tretinoína/administração & dosagem
15.
J Exp Med ; 212(5): 649-63, 2015 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-25870200

RESUMO

Hematopoietic stem cells (HSCs) emerge from aortic endothelium via the endothelial-to-hematopoietic transition (EHT). The molecular mechanisms that initiate and regulate EHT remain poorly understood. Here, we show that adenosine signaling regulates hematopoietic stem and progenitor cell (HSPC) development in zebrafish embryos. The adenosine receptor A2b is expressed in the vascular endothelium before HSPC emergence. Elevated adenosine levels increased runx1(+)/cmyb(+) HSPCs in the dorsal aorta, whereas blocking the adenosine pathway decreased HSPCs. Knockdown of A2b adenosine receptor disrupted scl(+) hemogenic vascular endothelium and the subsequent EHT process. A2b adenosine receptor activation induced CXCL8 via cAMP-protein kinase A (PKA) and mediated hematopoiesis. We further show that adenosine increased multipotent progenitors in a mouse embryonic stem cell colony-forming assay and in embryonic day 10.5 aorta-gonad-mesonephros explants. Our results demonstrate that adenosine signaling plays an evolutionary conserved role in the first steps of HSPC formation in vertebrates.


Assuntos
Adenosina/metabolismo , Aorta/metabolismo , Endotélio Vascular/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Receptor A2B de Adenosina/metabolismo , Adenosina/genética , Animais , Aorta/citologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Endotélio Vascular/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Camundongos , Camundongos Knockout , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Receptor A2B de Adenosina/genética
16.
Dev Cell ; 32(6): 756-64, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25752963

RESUMO

CRISPR/Cas9 technology of genome editing has greatly facilitated the targeted inactivation of genes in vitro and in vivo in a wide range of organisms. In zebrafish, it allows the rapid generation of knockout lines by simply injecting a guide RNA (gRNA) and Cas9 mRNA into one-cell stage embryos. Here, we report a simple and scalable CRISPR-based vector system for tissue-specific gene inactivation in zebrafish. As proof of principle, we used our vector with the gata1 promoter driving Cas9 expression to silence the urod gene, implicated in heme biosynthesis, specifically in the erythrocytic lineage. Urod targeting yielded red fluorescent erythrocytes in zebrafish embryos, recapitulating the phenotype observed in the yquem mutant. While F0 embryos displayed mosaic gene disruption, the phenotype appeared very penetrant in stable F1 fish. This vector system constitutes a unique tool to spatially control gene knockout and greatly broadens the scope of loss-of-function studies in zebrafish.


Assuntos
Técnicas de Inativação de Genes , Engenharia Genética/métodos , Peixe-Zebra/embriologia , Anemia de Diamond-Blackfan/genética , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Modelos Animais de Doenças , Eritrócitos/citologia , Vetores Genéticos , Proteínas Luminescentes/genética , Regiões Promotoras Genéticas/genética , Proteína Supressora de Tumor p53/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteína Vermelha Fluorescente
17.
Blood ; 124(25): 3772-80, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25258343

RESUMO

PML/RARA, a potent transcriptional inhibitor of nuclear receptor signaling, represses myeloid differentiation genes and drives acute promyelocytic leukemia (APL). Association of the retinoid X receptor-α (RXRA) coreceptor to PML/RARA is required for transformation, with RXRA promoting its efficient DNA binding. APL is exquisitely sensitive to retinoic acid (RA) and arsenic trioxide (arsenic), which both trigger cell differentiation in vivo. Whereas RA elicits transcriptional activation of PML/RARA targets, how arsenic triggers differentiation remains unclear. Here we demonstrate that extinction of PML/RARA triggers terminal differentiation in vivo. Similarly, ablation of retinoid X receptors loosens PML/RARA DNA binding, inducing terminal differentiation of APL cells ex vivo or in vivo. RXRA sumoylation directly contributes to PML/RARA-dependent transformation ex vivo, presumably by enhancing transcriptional repression. Thus, APL differentiation is a default program triggered by clearance of PML/RARA-bound promoters, rather than obligatory active transcriptional activation, explaining how arsenic elicits APL maturation through PML/RARA degradation.


Assuntos
Diferenciação Celular/genética , Leucemia Promielocítica Aguda/genética , Proteínas de Fusão Oncogênica/genética , Regiões Promotoras Genéticas/genética , Animais , Antineoplásicos/farmacologia , Trióxido de Arsênio , Arsenicais/farmacologia , Células COS , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Células Cultivadas , Chlorocebus aethiops , Perfilação da Expressão Gênica , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Nus , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/metabolismo , Óxidos/farmacologia , Ligação Proteica , Interferência de RNA , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sumoilação , Ativação Transcricional/genética , Tretinoína/farmacologia
18.
Int J Cancer ; 135(10): 2262-72, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25130873

RESUMO

Inevitably fatal some 40 years, acute promyelocytic leukemia (APL) can now be cured in more than 95% of cases. This clinical success story is tightly linked to tremendous progress in our understanding of retinoic acid (RA) signaling. The discovery of retinoic acid receptor alpha (RARA) was followed by the cloning of the chromosomal translocations driving APL, all of which involve RARA. Since then, new findings on the biology of nuclear receptors have progressively enlightened the basis for the clinical efficacy of RA in APL. Reciprocally, the disease offered a range of angles to approach the cellular and molecular mechanisms of RA action. This virtuous circle contributed to make APL one of the best-understood cancers from both clinical and biological standpoints. Yet, some important questions remain unanswered including how lessons learnt from RA-triggered APL cure can help design new therapies for other malignancies.


Assuntos
Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/terapia , Tretinoína/metabolismo , Humanos , Transdução de Sinais
19.
Cancer Res ; 74(8): 2328-39, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24566867

RESUMO

HDAC inhibitors (HDACi) are widely used in the clinic to sensitize tumorigenic cells for treatment with other anticancer compounds. The major drawback of HDACi is the broad inhibition of the plethora of HDAC-containing complexes. In acute promyelocytic leukemia (APL), repression by the PML-RARα oncofusion protein is mediated by an HDAC-containing complex that can be dissociated by pharmacologic doses of all trans retinoic acid (ATRA) inducing differentiation and cell death at the expense of side effects and recurrence. We hypothesized that the context-specific close physical proximity of a retinoid and HDACi-binding protein in the repressive PML-RARα-HDAC complex may permit selective targeting by a hybrid molecule of ATRA with a 2-aminoanilide tail of the HDAC inhibitor MS-275, yielding MC2392. We show that MC2392 elicits weak ATRA and essentially no HDACi activity in vitro or in vivo. Genome-wide epigenetic analyses revealed that in NB4 cells expressing PML-RARα, MC2392 induces changes in H3 acetylation at a small subset of PML-RARα-binding sites. RNA-seq reveals that MC2392 alters expression of a number of stress-responsive and apoptotic genes. Concordantly, MC2392 induced rapid and massive, caspase-8-dependent cell death accompanied by RIP1 induction and ROS production. Solid and leukemic tumors are not affected by MC2392, but expression of PML-RARα conveys efficient MC2392-induced cell death. Our data suggest a model in which MC2392 binds to the RARα moiety and selectively inhibits the HDACs resident in the repressive complex responsible for the transcriptional impairment in APLs. Our findings provide proof-of-principle of the concept of a context-dependent targeted therapy.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Retinoides/farmacologia , Acetilação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 8/metabolismo , Morte Celular , Diferenciação Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transfecção , Tretinoína/farmacologia , Células U937
20.
Nat Med ; 20(2): 167-74, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24412926

RESUMO

Acute promyelocytic leukemia (APL) is driven by the promyelocytic leukemia (PML)-retinoic acid receptor-α (PML-RARA) fusion protein, which interferes with nuclear receptor signaling and PML nuclear body (NB) assembly. APL is the only malignancy definitively cured by targeted therapies: retinoic acid (RA) and/or arsenic trioxide, which both trigger PML-RARA degradation through nonoverlapping pathways. Yet, the cellular and molecular determinants of treatment efficacy remain disputed. We demonstrate that a functional Pml-transformation-related protein 53 (Trp53) axis is required to eradicate leukemia-initiating cells in a mouse model of APL. Upon RA-induced PML-RARA degradation, normal Pml elicits NB reformation and induces a Trp53 response exhibiting features of senescence but not apoptosis, ultimately abrogating APL-initiating activity. Apart from triggering PML-RARA degradation, arsenic trioxide also targets normal PML to enhance NB reformation, which may explain its clinical potency, alone or with RA. This Pml-Trp53 checkpoint initiated by therapy-triggered NB restoration is specific for PML-RARA-driven APL, but not the RA-resistant promyelocytic leukemia zinc finger (PLZF)-RARA variant. Yet, as NB biogenesis is druggable, it could be therapeutically exploited in non-APL malignancies.


Assuntos
Leucemia Promielocítica Aguda/tratamento farmacológico , Proteínas Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Trióxido de Arsênio , Arsenicais/farmacologia , Biologia Computacional , Humanos , Estimativa de Kaplan-Meier , Leucemia Promielocítica Aguda/metabolismo , Camundongos , Análise em Microsséries , Óxidos/farmacologia , Proteína da Leucemia Promielocítica , Proteólise/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Receptor alfa de Ácido Retinoico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...