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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(7): 1223-1239.e20, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35290801

RESUMO

While CRISPR screens are helping uncover genes regulating many cell-intrinsic processes, existing approaches are suboptimal for identifying extracellular gene functions, particularly in the tissue context. Here, we developed an approach for spatial functional genomics called Perturb-map. We applied Perturb-map to knock out dozens of genes in parallel in a mouse model of lung cancer and simultaneously assessed how each knockout influenced tumor growth, histopathology, and immune composition. Moreover, we paired Perturb-map and spatial transcriptomics for unbiased analysis of CRISPR-edited tumors. We found that in Tgfbr2 knockout tumors, the tumor microenvironment (TME) was converted to a fibro-mucinous state, and T cells excluded, concomitant with upregulated TGFß and TGFß-mediated fibroblast activation, indicating that TGFß-receptor loss on cancer cells increased TGFß bioavailability and its immunosuppressive effects on the TME. These studies establish Perturb-map for functional genomics within the tissue at single-cell resolution with spatial architecture preserved and provide insight into how TGFß responsiveness of cancer cells can affect the TME.


Assuntos
Neoplasias , Microambiente Tumoral , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Genômica , Camundongos , Neoplasias/genética , Fator de Crescimento Transformador beta/genética
2.
Cell ; 175(4): 1141-1155.e16, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343902

RESUMO

CRISPR pools are being widely employed to identify gene functions. However, current technology, which utilizes DNA as barcodes, permits limited phenotyping and bulk-cell resolution. To enable novel screening capabilities, we developed a barcoding system operating at the protein level. We synthesized modules encoding triplet combinations of linear epitopes to generate >100 unique protein barcodes (Pro-Codes). Pro-Code-expressing vectors were introduced into cells and analyzed by CyTOF mass cytometry. Using just 14 antibodies, we detected 364 Pro-Code populations; establishing the largest set of protein-based reporters. By pairing each Pro-Code with a different CRISPR, we simultaneously analyzed multiple phenotypic markers, including phospho-signaling, on dozens of knockouts. Pro-Code/CRISPR screens found two interferon-stimulated genes, the immunoproteasome component Psmb8 and a chaperone Rtp4, are important for antigen-dependent immune editing of cancer cells and identified Socs1 as a negative regulator of Pd-l1. The Pro-Code technology enables simultaneous high-dimensional protein-level phenotyping of 100s of genes with single-cell resolution.


Assuntos
Sistemas CRISPR-Cas , Citometria de Fluxo/métodos , Genômica/métodos , Espectrometria de Massas/métodos , Análise de Célula Única/métodos , Animais , Epitopos/química , Epitopos/classificação , Epitopos/genética , Células HEK293 , Humanos , Imunofenotipagem/métodos , Células Jurkat , Camundongos Endogâmicos BALB C , Proteoma/química , Proteoma/classificação , Proteoma/genética , Células THP-1
3.
Cell ; 158(3): 506-21, 2014 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25083866

RESUMO

Adaptation of the endoplasmic reticulum (ER) pathway for MHC class I (MHC-I) presentation in dendritic cells enables cross-presentation of peptides derived from phagocytosed microbes, infected cells, or tumor cells to CD8 T cells. How these peptides intersect with MHC-I molecules remains poorly understood. Here, we show that MHC-I selectively accumulate within phagosomes carrying microbial components, which engage Toll-like receptor (TLR) signaling. Although cross-presentation requires Sec22b-mediated phagosomal recruitment of the peptide loading complex from the ER-Golgi intermediate compartment (ERGIC), this step is independent of TLR signaling and does not deliver MHC-I. Instead, MHC-I are recruited from an endosomal recycling compartment (ERC), which is marked by Rab11a, VAMP3/cellubrevin, and VAMP8/endobrevin and holds large reserves of MHC-I. While Rab11a activity stocks ERC stores with MHC-I, MyD88-dependent TLR signals drive IκB-kinase (IKK)2-mediated phosphorylation of phagosome-associated SNAP23. Phospho-SNAP23 stabilizes SNARE complexes orchestrating ERC-phagosome fusion, enrichment of phagosomes with ERC-derived MHC-I, and subsequent cross-presentation during infection.


Assuntos
Apresentação de Antígeno , Endossomos/metabolismo , Fagossomos/metabolismo , Receptores Toll-Like/metabolismo , Animais , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Tecido Linfoide , Camundongos , Ovalbumina/imunologia , Fagocitose , Fosforilação , Transporte Proteico , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Receptores Toll-Like/imunologia , Proteínas rab de Ligação ao GTP/metabolismo
4.
Immunity ; 48(2): 271-285.e5, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466757

RESUMO

Stem cells are critical for the maintenance of many tissues, but whether their integrity is maintained in the face of immunity is unclear. Here we found that cycling epithelial stem cells, including Lgr5+ intestinal stem cells, as well as ovary and mammary stem cells, were eliminated by activated T cells, but quiescent stem cells in the hair follicle and muscle were resistant to T cell killing. Immune evasion was an intrinsic property of the quiescent stem cells resulting from systemic downregulation of the antigen presentation machinery, including MHC class I and TAP proteins, and is mediated by the transactivator NLRC5. This process was reversed upon stem cell entry into the cell cycle. These studies identify a link between stem cell quiescence, antigen presentation, and immune evasion. As cancer-initiating cells can derive from stem cells, these findings may help explain how the earliest cancer cells evade immune surveillance.


Assuntos
Folículo Piloso/citologia , Evasão da Resposta Imune , Vigilância Imunológica , Células-Tronco/imunologia , Animais , Apresentação de Antígeno , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Receptores Acoplados a Proteínas G/fisiologia , Evasão Tumoral
5.
Nat Immunol ; 15(1): 54-62, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24270517

RESUMO

miR-126 is a microRNA expressed predominately by endothelial cells and controls angiogenesis. We found miR-126 was required for the innate response to pathogen-associated nucleic acids and that miR-126-deficient mice had greater susceptibility to infection with pseudotyped HIV. Profiling of miRNA indicated that miR-126 had high and specific expression by plasmacytoid dendritic cells (pDCs). Moreover, miR-126 controlled the survival and function of pDCs and regulated the expression of genes encoding molecules involved in the innate response, including Tlr7, Tlr9 and Nfkb1, as well as Kdr, which encodes the growth factor receptor VEGFR2. Deletion of Kdr in DCs resulted in reduced production of type I interferon, which supports the proposal of a role for VEGFR2 in miR-126 regulation of pDCs. Our studies identify the miR-126-VEGFR2 axis as an important regulator of the innate response that operates through multiscale control of pDCs.


Assuntos
Células Dendríticas/imunologia , Imunidade Inata/imunologia , MicroRNAs/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Células Dendríticas/metabolismo , Citometria de Fluxo , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Imunidade Inata/genética , Immunoblotting , Interferon-alfa/sangue , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/genética , MicroRNAs/metabolismo , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/imunologia , Subunidade p50 de NF-kappa B/metabolismo , Ácidos Nucleicos/imunologia , Ácidos Nucleicos/metabolismo , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/metabolismo , Transcriptoma/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Circ Res ; 123(6): 673-685, 2018 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-30355233

RESUMO

RATIONALE: Abnormal SUMOylation has emerged as a characteristic of heart failure (HF) pathology. Previously, we found reduced SUMO1 (small ubiquitin-like modifier 1) expression and SERCA2a (sarcoplasmic reticulum Ca2+-ATPase) SUMOylation in human and animal HF models. SUMO1 gene delivery or small molecule activation of SUMOylation restored SERCA2a SUMOylation and cardiac function in HF models. Despite the critical role of SUMO1 in HF, the regulatory mechanisms underlying SUMO1 expression are largely unknown. OBJECTIVE: To examine miR-146a-mediated SUMO1 regulation and its consequent effects on cardiac morphology and function. METHODS AND RESULTS: In this study, miR-146a was identified as a SUMO1-targeting microRNA in the heart. A strong correlation was observed between miR-146a and SUMO1 expression in failing mouse and human hearts. miR-146a was manipulated in cardiomyocytes through AAV9 (adeno-associated virus serotype 9)-mediated gene delivery, and cardiac morphology and function were analyzed by echocardiography and hemodynamics. Overexpression of miR-146a reduced SUMO1 expression, SERCA2a SUMOylation, and cardiac contractility in vitro and in vivo. The effects of miR-146a inhibition on HF pathophysiology were examined by transducing a tough decoy of miR-146a into mice subjected to transverse aortic constriction. miR-146a inhibition improved cardiac contractile function and normalized SUMO1 expression. The regulatory mechanisms of miR-146a upregulation were elucidated by examining the major miR-146a-producing cell types and transfer mechanisms. Notably, transdifferentiation of fibroblasts triggered miR-146a overexpression and secretion through extracellular vesicles, and the extracellular vesicle-associated miR-146a transfer was identified as the causative mechanism of miR-146a upregulation in failing cardiomyocytes. Finally, extracellular vesicles isolated from failing hearts were shown to contain high levels of miR-146a and exerted negative effects on the SUMO1/SERCA2a signaling axis and hence cardiomyocyte contractility. CONCLUSIONS: Taken together, our results show that miR-146a is a novel regulator of the SUMOylation machinery in the heart, which can be targeted for therapeutic intervention.


Assuntos
Cardiomegalia/metabolismo , Insuficiência Cardíaca/metabolismo , MicroRNAs/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Proteína SUMO-1/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Comunicação Celular , Transdiferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Fibroblastos/metabolismo , Fibroblastos/patologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Camundongos , MicroRNAs/genética , Miócitos Cardíacos/patologia , Proteína SUMO-1/genética , Transdução de Sinais , Sumoilação
7.
Nat Methods ; 9(8): 840-6, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22751203

RESUMO

We introduce two large-scale resources for functional analysis of microRNA (miRNA): a decoy library for inhibiting miRNA function and a sensor library for monitoring microRNA activity. To take advantage of the sensor library, we developed a high-throughput assay called Sensor-seq to simultaneously quantify the activity of hundreds of miRNAs. Using this approach, we show that only the most abundant miRNAs in a cell mediate target suppression. Over 60% of detected miRNAs had no discernible activity, which indicated that the functional 'miRNome' of a cell is considerably smaller than currently inferred from profiling studies. Moreover, some highly expressed miRNAs exhibited relatively weak activity, which in some cases correlated with a high target-to-miRNA ratio or increased nuclear localization of the miRNA. Finally, we show that the miRNA decoy library can be used for pooled loss-of-function studies. These tools are valuable resources for studying miRNA biology and for miRNA-based therapeutics.


Assuntos
Técnicas Biossensoriais , Biblioteca Gênica , Vetores Genéticos/genética , Ensaios de Triagem em Larga Escala , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Humanos , MicroRNAs/antagonistas & inibidores
8.
Cancer Immunol Res ; 10(11): 1354-1369, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36095236

RESUMO

Tumor-associated macrophages (TAM) are one of the most abundant cell types in many solid tumors and typically exert protumor effects. This has led to an interest in macrophage-depleting agents for cancer therapy, but approaches developed to date have had limited success in clinical trials. Here, we report the development of a strategy for TAM depletion in mouse solid tumor models using chimeric antigen receptor (CAR) T cells targeting the macrophage marker F4/80 (F4.CAR-T). F4.CAR-T cells effectively killed macrophages in vitro and in vivo without toxicity. When injected into mice bearing orthotopic lung tumors, F4.CAR-T cells infiltrated tumor lesions and delayed tumor growth comparably with PD-1 blockade, and significantly extended mouse survival. Antitumor effects were mediated by F4.CAR-T-produced IFNγ, which promoted upregulation of MHC molecules on cancer cells and tumor-infiltrating myeloid cells. Notably, F4.CAR-T promoted expansion of endogenous CD8 T cells specific for tumor-associated antigen and led to immune editing of highly antigenic tumor cell clones. Antitumor impact was also observed in mouse models of ovarian and pancreatic cancer. These studies provide proof of principle to support CAR T-cell targeting of TAMs as a means to enhance antitumor immunity.


Assuntos
Receptores de Antígenos Quiméricos , Linfócitos T , Animais , Camundongos , Antígenos de Neoplasias , Linhagem Celular Tumoral , Modelos Animais de Doenças , Imunoterapia Adotiva , Macrófagos/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Progressão da Doença
9.
Cancer Discov ; 11(3): 599-613, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33334730

RESUMO

T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.


Assuntos
Citotoxicidade Imunológica , Imunoterapia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Receptor fas/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Efeito Espectador/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Sistemas CRISPR-Cas , Modelos Animais de Doenças , Edição de Genes , Engenharia Genética , Humanos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Camundongos , Camundongos Knockout , Neoplasias/etiologia , Neoplasias/terapia , Receptores de Antígenos Quiméricos , Especificidade do Receptor de Antígeno de Linfócitos T , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Nat Med ; 27(5): 851-861, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33958797

RESUMO

Langerhans cell histiocytosis (LCH) is a potentially fatal condition characterized by granulomatous lesions with characteristic clonal mononuclear phagocytes (MNPs) harboring activating somatic mutations in mitogen-activated protein kinase (MAPK) pathway genes, most notably BRAFV600E. We recently discovered that the BRAFV600E mutation can also affect multipotent hematopoietic progenitor cells (HPCs) in multisystem LCH disease. How the BRAFV600E mutation in HPCs leads to LCH is not known. Here we show that enforced expression of the BRAFV600E mutation in early mouse and human multipotent HPCs induced a senescence program that led to HPC growth arrest, apoptosis resistance and a senescence-associated secretory phenotype (SASP). SASP, in turn, promoted HPC skewing toward the MNP lineage, leading to the accumulation of senescent MNPs in tissue and the formation of LCH lesions. Accordingly, elimination of senescent cells using INK-ATTAC transgenic mice, as well as pharmacologic blockade of SASP, improved LCH disease in mice. These results identify senescent cells as a new target for the treatment of LCH.


Assuntos
Senescência Celular/genética , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/patologia , Células de Langerhans/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Apoptose/genética , Proliferação de Células/genética , Senescência Celular/efeitos dos fármacos , Citocinas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
11.
Stem Cell Reports ; 12(1): 122-134, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30629937

RESUMO

Tumor-initiating cells (TICs) contribute to drug resistance and tumor recurrence in cancers, thus experimental approaches to dissect the complexity of TICs are required to design successful TIC therapeutic strategies. Here, we show that miRNA-3' UTR sensor vectors can be used as a pathway-based method to identify, enrich, and analyze TICs from primary solid tumor patient samples. We have found that an miR-181ahigh subpopulation of cells sorted from primary ovarian tumor cells exhibited TIC properties in vivo, were enriched in response to continuous cisplatin treatment, and showed activation of numerous major stem cell regulatory pathways. This miRNA-sensor-based platform enabled high-throughput drug screening leading to identification of BET inhibitors as transcriptional inhibitors of miR-181a. Taken together, we provide a valuable miRNA-sensor-based approach to broaden the understanding of complex TIC regulatory mechanisms in cancers and to identify miRNA-targeting drugs.


Assuntos
Antineoplásicos/farmacologia , Técnicas Biossensoriais/métodos , Descoberta de Drogas/métodos , MicroRNAs/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Regiões 3' não Traduzidas , Linhagem Celular Tumoral , Feminino , Humanos , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia
12.
J Exp Med ; 216(10): 2265-2281, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31350310

RESUMO

Microglia, the brain resident macrophages, critically shape forebrain neuronal circuits. However, their precise function in the cerebellum is unknown. Here we show that human and mouse cerebellar microglia express a unique molecular program distinct from forebrain microglia. Cerebellar microglial identity was driven by the CSF-1R ligand CSF-1, independently of the alternate CSF-1R ligand, IL-34. Accordingly, CSF-1 depletion from Nestin+ cells led to severe depletion and transcriptional alterations of cerebellar microglia, while microglia in the forebrain remained intact. Strikingly, CSF-1 deficiency and alteration of cerebellar microglia were associated with reduced Purkinje cells, altered neuronal function, and defects in motor learning and social novelty interactions. These findings reveal a novel CSF-1-CSF-1R signaling-mediated mechanism that contributes to motor function and social behavior.


Assuntos
Comportamento Animal/fisiologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Microglia/metabolismo , Atividade Motora/fisiologia , Células de Purkinje/metabolismo , Transdução de Sinais/fisiologia , Comportamento Social , Animais , Humanos , Fator Estimulador de Colônias de Macrófagos/genética , Camundongos , Camundongos Transgênicos , Células de Purkinje/citologia , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
13.
Cell Rep ; 13(3): 504-515, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26456833

RESUMO

Somatic PTPN11 mutations cause juvenile myelomonocytic leukemia (JMML). Germline PTPN11 defects cause Noonan syndrome (NS), and specific inherited mutations cause NS/JMML. Here, we report that hematopoietic cells differentiated from human induced pluripotent stem cells (hiPSCs) harboring NS/JMML-causing PTPN11 mutations recapitulated JMML features. hiPSC-derived NS/JMML myeloid cells exhibited increased signaling through STAT5 and upregulation of miR-223 and miR-15a. Similarly, miR-223 and miR-15a were upregulated in 11/19 JMML bone marrow mononuclear cells harboring PTPN11 mutations, but not those without PTPN11 defects. Reducing miR-223's function in NS/JMML hiPSCs normalized myelogenesis. MicroRNA target gene expression levels were reduced in hiPSC-derived myeloid cells as well as in JMML cells with PTPN11 mutations. Thus, studying an inherited human cancer syndrome with hiPSCs illuminated early oncogenesis prior to the accumulation of secondary genomic alterations, enabling us to discover microRNA dysregulation, establishing a genotype-phenotype association for JMML and providing therapeutic targets.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Leucemia Mielomonocítica Juvenil/metabolismo , Células Mieloides/citologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patologia , MicroRNAs/genética , Mutação , Células Mieloides/metabolismo , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Regulação para Cima
14.
Nat Commun ; 5: 2977, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24394555

RESUMO

Ovarian cancer is a leading cause of cancer deaths among women. Effective targets to treat advanced epithelial ovarian cancer (EOC) and biomarkers to predict treatment response are still lacking because of the complexity of pathways involved in ovarian cancer progression. Here we show that miR-181a promotes TGF-ß-mediated epithelial-to-mesenchymal transition via repression of its functional target, Smad7. miR-181a and phosphorylated Smad2 are enriched in recurrent compared with matched-primary ovarian tumours and their expression is associated with shorter time to recurrence and poor outcome in patients with EOC. Furthermore, ectopic expression of miR-181a results in increased cellular survival, migration, invasion, drug resistance and in vivo tumour burden and dissemination. In contrast, miR-181a inhibition via decoy vector suppression and Smad7 re-expression results in significant reversion of these phenotypes. Combined, our findings highlight an unappreciated role for miR-181a, Smad7, and the TGF-ß signalling pathway in high-grade serous ovarian cancer.


Assuntos
Carcinoma Endometrioide/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Císticas, Mucinosas e Serosas/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Proteína Smad2/metabolismo , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Idoso , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patologia , Carcinoma Epitelial do Ovário , Movimento Celular/genética , Sobrevivência Celular/genética , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Estadiamento de Neoplasias , Neoplasias Císticas, Mucinosas e Serosas/metabolismo , Neoplasias Císticas, Mucinosas e Serosas/patologia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação
15.
Curr Biol ; 21(5): 369-76, 2011 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21353554

RESUMO

Considerable details about microRNA (miRNA) biogenesis and regulation have been uncovered, but little is known about the fate of the miRNA subsequent to target regulation. To gain insight into this process, we carried out kinetic analysis of a miRNA's turnover following termination of its biogenesis and during regulation of a target that is not subject to Ago2-mediated catalytic cleavage. By quantitating the number of molecules of the miRNA and its target in steady state and in the course of its decay, we found that each miRNA molecule was able to regulate at least two target transcripts, providing in vivo evidence that the miRNA is not irreversibly sequestered with its target and that the nonslicing pathway of miRNA regulation is multiple-turnover. Using deep sequencing, we further show that miRNA recycling is limited by target regulation, which promotes posttranscriptional modifications to the 3' end of the miRNA and accelerates the miRNA's rate of decay. These studies provide new insight into the efficiency of miRNA regulation that help to explain how a miRNA can regulate a vast number of transcripts and that identify one of the mechanisms that impart specificity to miRNA decay in mammalian cells.


Assuntos
Regulação da Expressão Gênica/genética , MicroRNAs/genética , MicroRNAs/fisiologia , Estabilidade de RNA/fisiologia , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Citometria de Fluxo , Vetores Genéticos , Humanos , Cinética , Modelos Genéticos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Oligonucleotídeos/genética , Receptores de Fator de Crescimento Neural/genética , Análise de Sequência de DNA
16.
Methods Mol Biol ; 667: 215-33, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20827537

RESUMO

An essential requirement for discovering microRNAs that may be relevant to an immune cell's function is to identify the microRNAs that are active in the cell and the genes they target. As several chapters in this volume describe, there are a number of technologies available for profiling microRNA expression, including oligonucleotide array-based approaches, real-time PCR, and, now, deep-sequencing. A complementary approach to expression profiling is the use of a microRNA reporter vector for assaying microRNA activity. In their simplest form, these vectors are comprised of a reporter gene tethered to tandem repeats of a sequence that is complementary to a specific microRNA. This technology enables the activity of a microRNA to be detected, and at single-cell resolution, and provides a means to help identify microRNAs that may have a role in cell function. This is particularly relevant for studying microRNAs in the highly heterogeneous cellular network of the immune system. Reporter vectors have also proved useful for validating microRNA target sites and 3' untranslated regions (UTR) that are under microRNA control. This chapter describes how to construct, produce, and use a reporter vector for assaying microRNA activity, and for validating a microRNA target.


Assuntos
Perfilação da Expressão Gênica , Genes Reporter , MicroRNAs/genética , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Perfilação da Expressão Gênica/instrumentação , Perfilação da Expressão Gênica/métodos , Vetores Genéticos , Células HEK293 , Humanos , Sistema Imunitário/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos/instrumentação , Análise de Sequência com Séries de Oligonucleotídeos/métodos
17.
Mol Cancer Ther ; 8(9): 2684-92, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19723889

RESUMO

Alterations in the human 13q14 genomic region containing microRNAs mir-15a and mir-16-1 are present in most human chronic lymphocytic leukemia (CLL). We have previously found the development of CLL in the New Zealand Black murine model to be associated with a point mutation in the primary mir-15a/16-1 region, which correlated with a decrease in mature miR-16 and miR-15a levels. In this study, addition of exogenous miR-15a and miR-16 led to an accumulation of cells in G(1) in non-New Zealand Black B cell and New Zealand Black-derived malignant B-1 cell lines. However, the New Zealand Black line had significantly greater G(1) accumulation, suggesting a restoration of cell cycle control upon exogenous miR-15a/16 addition. Our experiments showed a reduction in protein levels of cyclin D1, a miR-15a/16 target and cell cycle regulator of G(1)/S transition, in the New Zealand Black cell line following miR-15a/16 addition. These microRNAs were shown to directly target the cyclin D1 3' untranslated region using a green fluorescent protein lentiviral expression system. miR-16 was also shown to augment apoptosis induction by nutlin, a mouse double minute 2 (MDM2) antagonist, and genistein, a tyrosine kinase inhibitor, when added to a B-1 cell line derived from multiple in vivo passages of malignant B-1 cells from New Zealand Black mice with CLL. miR-16 synergized with nutlin and genistein to induce apoptosis. Our data support a role for the mir-15a/16-1 cluster in cell cycle regulation and suggest that these mature microRNAs in both the New Zealand Black model and human CLL may be targets for therapeutic efficacy in this disease.


Assuntos
Modelos Animais de Doenças , Leucemia Linfocítica Crônica de Células B/genética , MicroRNAs/genética , Animais , Antineoplásicos/farmacologia , Apoptose , Sequência de Bases , Ciclina D1/genética , Primers do DNA , Ensaios de Seleção de Medicamentos Antitumorais , Genisteína/uso terapêutico , Imidazóis/uso terapêutico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos Endogâmicos C57BL , Piperazinas/uso terapêutico , RNA Mensageiro/genética
18.
Nat Biotechnol ; 25(12): 1457-67, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18026085

RESUMO

We have shown previously that transgene expression can be suppressed in hematopoietic cells using vectors that are responsive to microRNA (miRNA) regulation. Here we investigate the potential of this approach for more sophisticated control of transgene expression. Analysis of the relationship between miRNA expression levels and target mRNA suppression suggested that suppression depends on a threshold miRNA concentration. Using this information, we generated vectors that rapidly adjust transgene expression in response to changes in miRNA expression. These vectors sharply segregated transgene expression between closely related states of therapeutically relevant cells, including dendritic cells, hematopoietic and embryonic stem cells, and their progeny, allowing positive/negative selection according to the cells' differentiation state. Moreover, two miRNA target sites were combined to restrict transgene expression to a specific cell type in the liver. Notably, the vectors did not detectably perturb endogenous miRNA expression or regulation of natural targets. The properties of miRNA-regulated vectors should allow for safer and more effective therapeutic applications.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Inativação Gênica/fisiologia , Marcação de Genes/métodos , MicroRNAs/genética , Células-Tronco/citologia , Células-Tronco/fisiologia , Transgenes/genética , Animais , Diferenciação Celular/genética , Humanos
19.
Mol Cell ; 18(4): 447-59, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15893728

RESUMO

The transcriptional coactivator Yes-associated protein (YAP) has been shown to interact with and to enhance p73-dependent apoptosis in response to DNA damage. Here, we show that YAP requires the promyelocytic leukemia gene (PML) and nuclear body localization to coactivate p73. YAP imparts selectivity to p73 by promoting the activation of a subset of p53 and/or p73 target promoters. Endogenous p73, YAP, and p300 proteins are concomitantly recruited onto the regulatory regions of the apoptotic target gene p53AIP1 only when cells are exposed to apoptotic conditions. Silencing of YAP by specific siRNA impairs p300 recruitment and reduces histone acetylation on the p53AIP1 target gene, resulting in delayed or reduced apoptosis mediated by p73. We also found that YAP contributes to the DNA damage-induced accumulation of p73 and potentiates the p300-mediated acetylation of p73. Altogether, our findings identify YAP as a key determinant of p73 gene targeting in response to DNA damage.


Assuntos
Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Acetilação , Antibióticos Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose , Proteínas de Ciclo Celular , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Genes Reporter , Genes Supressores de Tumor , Humanos , Proteínas de Neoplasias/metabolismo , Proteína da Leucemia Promielocítica , Proteínas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Proteína Tumoral p73 , Proteínas Supressoras de Tumor
20.
J Biol Chem ; 277(21): 18817-26, 2002 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-11893750

RESUMO

The p53 tumor suppressor gene is the most frequent target for genetic alterations in human cancers, whereas the recently discovered homologues p73 and p63 are rarely mutated. We and others have previously reported that human tumor-derived p53 mutants can engage in a physical association with different isoforms of p73, inhibiting their transcriptional activity. Here, we report that human tumor-derived p53 mutants can associate in vitro and in vivo with p63 through their respective core domains. We show that the interaction with mutant p53 impairs in vitro and in vivo sequence-specific DNA binding of p63 and consequently affects its transcriptional activity. We also report that in cells carrying endogenous mutant p53, such as T47D cells, p63 is unable to recruit some of its target gene promoters. Unlike wild-type p53, the binding to specific p53 mutants markedly counteracts p63-induced growth inhibition. This effect is, at least partially, mediated by the core domain of mutant p53. Thus, inactivation of p53 family members may contribute to the biological properties of specific p53 mutants in promoting tumorigenesis and in conferring selective survival advantage to cancer cells.


Assuntos
Genes Supressores de Tumor , Proteínas de Membrana , Mutação , Fosfoproteínas/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Sequência de Bases , Sítios de Ligação , DNA/metabolismo , Primers do DNA , Proteínas de Ligação a DNA , Histidina/metabolismo , Humanos , Fosfoproteínas/metabolismo , Ligação Proteica , Transativadores/metabolismo , Fatores de Transcrição , Triptofano/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA