Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 630(8016): 412-420, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38839950

RESUMO

The processes that govern human haematopoietic stem cell (HSC) self-renewal and engraftment are poorly understood and challenging to recapitulate in culture to reliably expand functional HSCs1-3. Here we identify MYC target 1 (MYCT1; also known as MTLC) as a crucial human HSC regulator that moderates endocytosis and environmental sensing in HSCs. MYCT1 is selectively expressed in undifferentiated human haematopoietic stem and progenitor cells (HSPCs) and endothelial cells but becomes markedly downregulated during HSC culture. Lentivirus-mediated knockdown of MYCT1 prevented human fetal liver and cord blood (CB) HSPC expansion and engraftment. By contrast, restoring MYCT1 expression improved the expansion and engraftment of cultured CB HSPCs. Single-cell RNA sequencing of human CB HSPCs in which MYCT1 was knocked down or overexpressed revealed that MYCT1 governs important regulatory programmes and cellular properties essential for HSC stemness, such as ETS factor expression and low mitochondrial activity. MYCT1 is localized in the endosomal membrane in HSPCs and interacts with vesicle trafficking regulators and signalling machinery. MYCT1 loss in HSPCs led to excessive endocytosis and hyperactive signalling responses, whereas restoring MYCT1 expression balanced culture-induced endocytosis and dysregulated signalling. Moreover, sorting cultured CB HSPCs on the basis of lowest endocytosis rate identified HSPCs with preserved MYCT1 expression and MYCT1-regulated HSC stemness programmes. Our work identifies MYCT1-moderated endocytosis and environmental sensing as essential regulatory mechanisms required to preserve human HSC stemness. Our data also pinpoint silencing of MYCT1 as a cell-culture-induced vulnerability that compromises human HSC expansion.


Assuntos
Autorrenovação Celular , Células-Tronco Hematopoéticas , Proteínas Nucleares , Animais , Feminino , Humanos , Masculino , Camundongos , Células Cultivadas , Endocitose , Endossomos/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Sangue Fetal/citologia , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Fígado/citologia , Fígado/metabolismo , Fígado/embriologia , Mitocôndrias/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Análise da Expressão Gênica de Célula Única
2.
Nat Commun ; 15(1): 1579, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383676

RESUMO

Oncogene-induced replication stress is a crucial driver of genomic instability and one of the key events contributing to the onset and evolution of cancer. Despite its critical role in cancer, the mechanisms that generate oncogene-induced replication stress remain not fully understood. Here, we report that an oncogenic c-Myc-dependent increase in cohesins on DNA contributes to the induction of replication stress. Accumulation of cohesins on chromatin is not sufficient to cause replication stress, but also requires cohesins to accumulate at specific sites in a CTCF-dependent manner. We propose that the increased accumulation of cohesins at CTCF site interferes with the progression of replication forks, contributing to oncogene-induced replication stress. This is different from, and independent of, previously suggested mechanisms of oncogene-induced replication stress. This, together with the reported protective role of cohesins in preventing replication stress-induced DNA damage, supports a double-edge involvement of cohesins in causing and tolerating oncogene-induced replication stress.


Assuntos
Coesinas , Neoplasias , Humanos , Cromatina , Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , DNA
3.
Blood ; 142(6): 519-532, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37339578

RESUMO

Developmental hematopoiesis consists of multiple, partially overlapping hematopoietic waves that generate the differentiated blood cells required for embryonic development while establishing a pool of undifferentiated hematopoietic stem cells (HSCs) for postnatal life. This multilayered design in which active hematopoiesis migrates through diverse extra and intraembryonic tissues has made it difficult to define a roadmap for generating HSCs vs non-self-renewing progenitors, especially in humans. Recent single-cell studies have helped in identifying the rare human HSCs at stages when functional assays are unsuitable for distinguishing them from progenitors. This approach has made it possible to track the origin of human HSCs to the unique type of arterial endothelium in the aorta-gonad-mesonephros region and document novel benchmarks for HSC migration and maturation in the conceptus. These studies have delivered new insights into the intricate process of HSC generation and provided tools to inform the in vitro efforts to replicate the physiological developmental journey from pluripotent stem cells via distinct mesodermal and endothelial intermediates to HSCs.


Assuntos
Embrião de Mamíferos , Células-Tronco Hematopoéticas , Feminino , Gravidez , Humanos , Hematopoese/fisiologia , Mesonefro
4.
Nature ; 604(7906): 534-540, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35418685

RESUMO

The ontogeny of human haematopoietic stem cells (HSCs) is poorly defined owing to the inability to identify HSCs as they emerge and mature at different haematopoietic sites1. Here we created a single-cell transcriptome map of human haematopoietic tissues from the first trimester to birth and found that the HSC signature RUNX1+HOXA9+MLLT3+MECOM+HLF+SPINK2+ distinguishes HSCs from progenitors throughout gestation. In addition to the aorta-gonad-mesonephros region, nascent HSCs populated the placenta and yolk sac before colonizing the liver at 6 weeks. A comparison of HSCs at different maturation stages revealed the establishment of HSC transcription factor machinery after the emergence of HSCs, whereas their surface phenotype evolved throughout development. The HSC transition to the liver marked a molecular shift evidenced by suppression of surface antigens reflecting nascent HSC identity, and acquisition of the HSC maturity markers CD133 (encoded by PROM1) and HLA-DR. HSC origin was tracked to ALDH1A1+KCNK17+ haemogenic endothelial cells, which arose from an IL33+ALDH1A1+ arterial endothelial subset termed pre-haemogenic endothelial cells. Using spatial transcriptomics and immunofluorescence, we visualized this process in ventrally located intra-aortic haematopoietic clusters. The in vivo map of human HSC ontogeny validated the generation of aorta-gonad-mesonephros-like definitive haematopoietic stem and progenitor cells from human pluripotent stem cells, and serves as a guide to improve their maturation to functional HSCs.


Assuntos
Células Endoteliais , Células-Tronco Hematopoéticas , Diferenciação Celular , Endotélio , Feminino , Hematopoese , Humanos , Mesonefro , Gravidez
5.
Nature ; 576(7786): 281-286, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31776511

RESUMO

Limited knowledge of the mechanisms that govern the self-renewal of human haematopoietic stem cells (HSCs), and why this fails in culture, have impeded the expansion of HSCs for transplantation1. Here we identify MLLT3 (also known as AF9) as a crucial regulator of HSCs that is highly enriched in human fetal, neonatal and adult HSCs, but downregulated in culture. Depletion of MLLT3 prevented the maintenance of transplantable human haematopoietic stem or progenitor cells (HSPCs) in culture, whereas stabilizing MLLT3 expression in culture enabled more than 12-fold expansion of transplantable HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Similar to endogenous MLLT3, overexpressed MLLT3 localized to active promoters in HSPCs, sustained levels of H3K79me2 and protected the HSC transcriptional program in culture. MLLT3 thus acts as HSC maintenance factor that links histone reader and modifying activities to modulate HSC gene expression, and may provide a promising approach to expand HSCs for transplantation.


Assuntos
Autorrenovação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Proteínas Nucleares/genética , Ligação Proteica
6.
Exp Hematol ; 65: 1-16, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29981365

RESUMO

Hematopoietic stem cells (HSCs) are multipotent cells responsible for the maintenance of the hematopoietic system throughout life. Dysregulation of the balance in HSC self-renewal, death, and differentiation can have serious consequences such as myelodysplastic syndromes or leukemia. All-trans retinoic acid (ATRA), the biologically active metabolite of vitamin A/RA, has been shown to have pleiotropic effects on hematopoietic cells, enhancing HSC self-renewal while also increasing differentiation of more mature progenitors. Furthermore, ATRA has been shown to have key roles in regulating the specification and formation of hematopoietic cells from pluripotent stem cells including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Here, we summarize the known roles of vitamin A and RA receptors in the regulation of hematopoiesis from HSCs, ES, and iPSCs.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes/citologia , Retinoides/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Humanos , Modelos Biológicos , Receptores do Ácido Retinoico/fisiologia , Transdução de Sinais
7.
Nat Biotechnol ; 34(11): 1168-1179, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27748754

RESUMO

The ability to generate hematopoietic stem cells from human pluripotent cells would enable many biomedical applications. We find that hematopoietic CD34+ cells in spin embryoid bodies derived from human embryonic stem cells (hESCs) lack HOXA expression compared with repopulation-competent human cord blood CD34+ cells, indicating incorrect mesoderm patterning. Using reporter hESC lines to track the endothelial (SOX17) to hematopoietic (RUNX1C) transition that occurs in development, we show that simultaneous modulation of WNT and ACTIVIN signaling yields CD34+ hematopoietic cells with HOXA expression that more closely resembles that of cord blood. The cultures generate a network of aorta-like SOX17+ vessels from which RUNX1C+ blood cells emerge, similar to hematopoiesis in the aorta-gonad-mesonephros (AGM). Nascent CD34+ hematopoietic cells and corresponding cells sorted from human AGM show similar expression of cell surface receptors, signaling molecules and transcription factors. Our findings provide an approach to mimic in vitro a key early stage in human hematopoiesis for the generation of AGM-derived hematopoietic lineages from hESCs.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Mesonefro/citologia , Mesonefro/embriologia , Neovascularização Fisiológica/fisiologia , Aorta/citologia , Aorta/embriologia , Aorta/crescimento & desenvolvimento , Diferenciação Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Gônadas/citologia , Gônadas/embriologia , Gônadas/crescimento & desenvolvimento , Células-Tronco Hematopoéticas/fisiologia , Humanos , Mesonefro/crescimento & desenvolvimento
8.
Nat Cell Biol ; 18(6): 595-606, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27183470

RESUMO

Pluripotent stem cells (PSCs) may provide a potential source of haematopoietic stem/progenitor cells (HSPCs) for transplantation; however, unknown molecular barriers prevent the self-renewal of PSC-HSPCs. Using two-step differentiation, human embryonic stem cells (hESCs) differentiated in vitro into multipotent haematopoietic cells that had the CD34(+)CD38(-/lo)CD90(+)CD45(+)GPI-80(+) fetal liver (FL) HSPC immunophenotype, but exhibited poor expansion potential and engraftment ability. Transcriptome analysis of immunophenotypic hESC-HSPCs revealed that, despite their molecular resemblance to FL-HSPCs, medial HOXA genes remained suppressed. Knockdown of HOXA7 disrupted FL-HSPC function and caused transcriptome dysregulation that resembled hESC-derived progenitors. Overexpression of medial HOXA genes prolonged FL-HSPC maintenance but was insufficient to confer self-renewal to hESC-HSPCs. Stimulation of retinoic acid signalling during endothelial-to-haematopoietic transition induced the HOXA cluster and other HSC/definitive haemogenic endothelium genes, and prolonged HSPC maintenance in culture. Thus, medial HOXA gene expression induced by retinoic acid signalling marks the establishment of the definitive HSPC fate and controls HSPC identity and function.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula , Genes Homeobox/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/citologia , Antígenos CD34/metabolismo , Células Cultivadas , Perfilação da Expressão Gênica/métodos , Proteínas de Homeodomínio/genética , Humanos , Antígenos Comuns de Leucócito/metabolismo , Transcriptoma
9.
Dev Cell ; 36(5): 479-80, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26954540

RESUMO

Reporting in Developmental Cell, Pereira et al. (2016) use in vitro lineage reprogramming insights to inform understanding of hematopoietic stem cell (HSC) development in vivo. They find Prom1(+)Sca1(+)CD34(+)CD45(-) hemogenic precursors, akin to fibroblast-derived hemato-vascular precursors, in mouse placenta and embryo. The cells mature into transplantable HSCs in culture.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Reprogramação Celular , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Embrionárias Murinas/citologia , Animais , Feminino , Gravidez
10.
Oncotarget ; 6(8): 5903-17, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25788273

RESUMO

Sporadic colorectal cancer (CRC) insurgence and progression depend on the activation of Wnt/ß-catenin signaling. Dickkopf (DKK)-1 is an extracellular inhibitor of Wnt/ß-catenin signaling that also has undefined ß-catenin-independent actions. Here we report for the first time that a proportion of DKK-1 locates within the nucleus of healthy small intestine and colon mucosa, and of CRC cells at specific chromatin sites of active transcription. Moreover, we show that DKK-1 regulates several cancer-related genes including the cancer stem cell marker aldehyde dehydrogenase 1A1 (ALDH1A1) and Ral-binding protein 1-associated Eps domain-containing 2 (REPS2), which are involved in detoxification of chemotherapeutic agents. Nuclear DKK-1 expression is lost along CRC progression; however, it remains high in a subset (15%) of CRC patients (n = 699) and associates with decreased progression-free survival (PFS) after chemotherapy administration and overall survival (OS) [adjusted HR, 1.65; 95% confidence interval (CI), 1.23-2.21; P = 0.002)]. Overexpression of ALDH1A1 and REPS2 associates with nuclear DKK-1 expression in tumors and correlates with decreased OS (P = 0.001 and 0.014) and PFS. In summary, our findings demonstrate a novel location of DKK-1 within the cell nucleus and support a role of nuclear DKK-1 as a predictive biomarker of chemoresistance in colorectal cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Aldeído Desidrogenase/biossíntese , Aldeído Desidrogenase/genética , Família Aldeído Desidrogenase 1 , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mucosa Intestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Prognóstico , Retinal Desidrogenase , Transdução de Sinais
11.
Cell Stem Cell ; 16(1): 80-7, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25465114

RESUMO

Advances in pluripotent stem cell and reprogramming technologies have given us the hope of generating hematopoietic stem cells (HSCs) in culture. To succeed, greater understanding of the self-renewing HSC during human development is required. We discovered that the glycophosphatidylinositol-anchored surface protein GPI-80 defines a subpopulation of human fetal liver hematopoietic stem/progenitor cells (HSPCs) with self-renewal ability. CD34(+)CD38(lo/-)CD90(+)GPI-80(+) HSPCs were the sole population that maintained proliferative potential and an undifferentiated state in stroma coculture and engrafted in immunodeficient mice. GPI-80 expression also enabled tracking of HSPCs once they emerged from endothelium and migrated between human fetal hematopoietic niches. GPI-80 colocalized on the surface of HSPCs with Integrin alpha-M (ITGAM), which in leukocytes cooperates with GPI-80 to support migration. Knockdown of GPI-80 or ITGAM was sufficient to compromise HSPC expansion in culture and engraftment in vivo. These findings indicate that human fetal HSCs employ mechanisms used in leukocyte adhesion and migration to mediate HSC self-renewal.


Assuntos
Amidoidrolases/metabolismo , Moléculas de Adesão Celular/metabolismo , Desenvolvimento Embrionário , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Antígeno CD11b/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Feto/citologia , Citometria de Fluxo , Proteínas Ligadas por GPI/metabolismo , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunofenotipagem , Fígado/citologia , Fígado/embriologia
12.
EMBO J ; 33(6): 534-5, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24562387

RESUMO

Stem cells ensure the maintenance of tissue homeostasis throughout life by tightly regulating their self-renewal and differentiation. In a recent study published in Nature, Nakada et al, 2014 unveil an unexpected endocrine mechanism that regulates hematopoietic stem cell (HSC) self-renewal.


Assuntos
Estrogênios/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Feminino , Masculino , Gravidez
13.
Cancer Cell ; 24(1): 15-29, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23845441

RESUMO

We show that BRAF(V600E) initiates an alternative pathway to colorectal cancer (CRC), which progresses through a hyperplasia/adenoma/carcinoma sequence. This pathway underlies significant subsets of CRCs with distinctive pathomorphologic/genetic/epidemiologic/clinical characteristics. Genetic and functional analyses in mice revealed a series of stage-specific molecular alterations driving different phases of tumor evolution and uncovered mechanisms underlying this stage specificity. We further demonstrate dose-dependent effects of oncogenic signaling, with physiologic Braf(V600E) expression being sufficient for hyperplasia induction, but later stage intensified Mapk-signaling driving both tumor progression and activation of intrinsic tumor suppression. Such phenomena explain, for example, the inability of p53 to restrain tumor initiation as well as its importance in invasiveness control, and the late stage specificity of its somatic mutation. Finally, systematic drug screening revealed sensitivity of this CRC subtype to targeted therapeutics, including Mek or combinatorial PI3K/Braf inhibition.


Assuntos
Neoplasias Colorretais/etiologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Transformação Celular Neoplásica , Neoplasias Colorretais/tratamento farmacológico , Inibidor p16 de Quinase Dependente de Ciclina , Ensaios de Seleção de Medicamentos Antitumorais , Sistema de Sinalização das MAP Quinases , Camundongos , Instabilidade de Microssatélites , Invasividade Neoplásica , Proteínas de Neoplasias/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteína Supressora de Tumor p53/fisiologia , Via de Sinalização Wnt
14.
Cell Cycle ; 12(3): 452-62, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23255218

RESUMO

The therapeutic potential of pharmacologic inhibition of bromodomain and extraterminal (BET) proteins has recently emerged in hematological malignancies and chronic inflammation. We find that BET inhibitor compounds (JQ1, I-Bet, I-Bet151 and MS417) reactivate HIV from latency. This is evident in polyclonal Jurkat cell populations containing latent infectious HIV, as well as in a primary T-cell model of HIV latency. Importantly, we show that this activation is dependent on the positive transcription elongation factor p-TEFb but independent from the viral Tat protein, arguing against the possibility that removal of the BET protein BRD4, which functions as a cellular competitor for Tat, serves as a primary mechanism for BET inhibitor action. Instead, we find that the related BET protein, BRD2, enforces HIV latency in the absence of Tat, pointing to a new target for BET inhibitor treatment in HIV infection. In shRNA-mediated knockdown experiments, knockdown of BRD2 activates HIV transcription to the same extent as JQ1 treatment, while a lesser effect is observed with BRD4. In single-cell time-lapse fluorescence microscopy, quantitative analyses across ~2,000 viral integration sites confirm the Tat-independent effect of JQ1 and point to positive effects of JQ1 on transcription elongation, while delaying re-initiation of the polymerase complex at the viral promoter. Collectively, our results identify BRD2 as a new Tat-independent suppressor of HIV transcription in latently infected cells and underscore the therapeutic potential of BET inhibitors in the reversal of HIV latency.


Assuntos
Infecções por HIV/virologia , HIV-1/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Azepinas/farmacologia , Benzodiazepinas/farmacologia , Linfócitos T CD4-Positivos/virologia , Proteínas de Ciclo Celular , Células Cultivadas , Células HEK293 , HIV-1/efeitos dos fármacos , HIV-1/genética , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Células Jurkat , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Fator B de Elongação Transcricional Positiva/genética , Fator B de Elongação Transcricional Positiva/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Triazóis/farmacologia , Latência Viral , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
15.
Proc Natl Acad Sci U S A ; 109(28): 11318-23, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22733740

RESUMO

PIK3R2 encodes a ubiquitous regulatory subunit (p85ß) of PI3K, an enzyme that generates 3-polyphosphoinositides at the plasma membrane. PI3K activation triggers cell survival and migration. We found that p85ß expression is elevated in breast and colon carcinomas and that its increased expression correlates with PI3K pathway activation and tumor progression. p85ß expression induced moderate PIP(3) generation at the cell membrane and enhanced cell invasion. In accordance, genetic alteration of pik3r2 expression levels modulated tumor progression in vivo. Increased p85ß expression thus represents a cellular strategy in cancer progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Neoplasias do Colo/metabolismo , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos SCID , Camundongos Transgênicos , Células NIH 3T3 , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Estrutura Terciária de Proteína , Transdução de Sinais
16.
Eur J Cancer ; 48(14): 2270-81, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22244828

RESUMO

Altered promoter DNA methylation, one of the most important molecular alterations in cancer, is proposed to correlate with deregulation of DNA methyltransferases, although the molecular mechanisms implicated are still poorly understood. Here we show that the de novo DNA methyltransferase DNMT3B is frequently repressed in human colorectal cancer cell lines (CCL) and primary tumours by aberrant DNA hypermethylation of its distal promoter. At the epigenome level, DNMT3B promoter hypermethylation was associated with the hypomethylation of gene promoters usually hypermethylated in the healthy colon. Forced DNMT3B overexpression in cancer cells restored the methylation levels of these promoters in the healthy colon. Our results show a new molecular mechanism of aberrant DNMT3B regulation in colon cancer and suggest that its expression is associated with the methylation of constitutively hypermethylated promoters in the healthy colon.


Assuntos
Neoplasias Colorretais/genética , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Regiões Promotoras Genéticas , Adulto , Idoso , Idoso de 80 Anos ou mais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias Colorretais/enzimologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Metilação de DNA/efeitos dos fármacos , Decitabina , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Células HCT116 , Células HT29 , Humanos , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas/efeitos dos fármacos , Transfecção , DNA Metiltransferase 3B
17.
Nucleic Acids Res ; 40(1): 116-31, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21911366

RESUMO

Global mechanisms defining the gene expression programs specific for hematopoiesis are still not fully understood. Here, we show that promoter DNA demethylation is associated with the activation of hematopoietic-specific genes. Using genome-wide promoter methylation arrays, we identified 694 hematopoietic-specific genes repressed by promoter DNA methylation in human embryonic stem cells and whose loss of methylation in hematopoietic can be associated with gene expression. The association between promoter methylation and gene expression was studied for many hematopoietic-specific genes including CD45, CD34, CD28, CD19, the T cell receptor (TCR), the MHC class II gene HLA-DR, perforin 1 and the phosphoinositide 3-kinase (PI3K) and results indicated that DNA demethylation was not always sufficient for gene activation. Promoter demethylation occurred either early during embryonic development or later on during hematopoietic differentiation. Analysis of the genome-wide promoter methylation status of induced pluripotent stem cells (iPSCs) generated from somatic CD34(+) HSPCs and differentiated derivatives from CD34(+) HSPCs confirmed the role of DNA methylation in regulating the expression of genes of the hemato-immune system, and indicated that promoter methylation of these genes may be associated to stemness. Together, these data suggest that promoter DNA demethylation might play a role in the tissue/cell-specific genome-wide gene regulation within the hematopoietic compartment.


Assuntos
Metilação de DNA , Regulação da Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Regiões Promotoras Genéticas , Animais , Desdiferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Hematopoéticas/citologia , Humanos , Recém-Nascido , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
18.
Aging (Albany NY) ; 3(2): 162-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21307403

RESUMO

Sirtuin 1 acts in various cell processes, deacetylating both chromatin and non-histone proteins, and its role in cancer and aging has long been studied and debated. Here we discuss another aspect of SirT1 biology, its function as a stem cell pluripotency and differentiation regulator. We evaluate the implications of these findings in sirtuin inhibition-based cancer treatment and in the application of sirtuin activation for anti-aging therapy.


Assuntos
Envelhecimento/fisiologia , Neoplasias/fisiopatologia , Sirtuína 1/metabolismo , Animais , Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células-Tronco/fisiologia
19.
Mol Cancer ; 9: 170, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20591152

RESUMO

BACKGROUND: Wnt factors control cell differentiation through semi-independent molecular cascades known as the beta-catenin-dependent (canonical) and -independent (non-canonical) Wnt signalling pathways. Genetic and epigenetic alteration of components of the canonical Wnt signalling pathway is one of the primary mechanisms underlying colon cancer. Despite increasing evidence of the role of the non-canonical pathways in tumourigenesis, however, the underlying molecular mechanisms are poorly understood. RESULTS: Here we report that the receptor tyrosine kinase-like orphan receptor 2 (ROR2), a transmembrane receptor for Wnt factors that activates non-canonical pathways, is frequently repressed by aberrant promoter hypermethylation in human colon cancer cell lines and primary tumours. By restoring ROR2 activity in colon cancer cells harbouring ROR2 promoter hypermethylation, we show that the role of ROR2 in colon cancer cells is mediated, at least in part, by canonical Wnt and that its epigenetic-dependent loss can be pro-tumourigenic. CONCLUSIONS: Our data show the importance of epigenetic alterations of ROR2 in colon cancer, highlighting the close interconnection between canonical and non-canonical Wnt signalling pathways in this type of tumour.


Assuntos
Neoplasias do Colo/genética , Epigênese Genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Proteínas Wnt/fisiologia , Neoplasias do Colo/patologia , Metilação de DNA , Humanos , Regiões Promotoras Genéticas
20.
PLoS One ; 5(4): e10192, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20419139

RESUMO

BACKGROUND: Human embryonic stem cells (hESCs) are an attractive resource for new therapeutic approaches that involve tissue regeneration. hESCs have exhibited low immunogenicity due to low levels of Mayor Histocompatibility Complex (MHC) class-I and absence of MHC class-II expression. Nevertheless, the mechanisms regulating MHC expression in hESCs had not been explored. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed the expression levels of classical and non-classical MHC class-I, MHC class-II molecules, antigen-processing machinery (APM) components and NKG2D ligands (NKG2D-L) in hESCs, induced pluripotent stem cells (iPSCs) and NTera2 (NT2) teratocarcinoma cell line. Epigenetic mechanisms involved in the regulation of these genes were investigated by bisulfite sequencing and chromatin immunoprecipitation (ChIP) assays. We showed that low levels of MHC class-I molecules were associated with absent or reduced expression of the transporter associated with antigen processing 1 (TAP-1) and tapasin (TPN) components in hESCs and iPSCs, which are involved in the transport and load of peptides. Furthermore, lack of beta2-microglobulin (beta2m) light chain in these cells limited the expression of MHC class I trimeric molecule on the cell surface. NKG2D ligands (MICA, MICB) were observed in all pluripotent stem cells lines. Epigenetic analysis showed that H3K9me3 repressed the TPN gene in undifferentiated cells whilst HLA-B and beta2m acquired the H3K4me3 modification during the differentiation to embryoid bodies (EBs). Absence of HLA-DR and HLA-G expression was regulated by DNA methylation. CONCLUSIONS/SIGNIFICANCE: Our data provide fundamental evidence for the epigenetic control of MHC in hESCs and iPSCs. Reduced MHC class I and class II expression in hESCs and iPSCs can limit their recognition by the immune response against these cells. The knowledge of these mechanisms will further allow the development of strategies to induce tolerance and improve stem cell allograft acceptance.


Assuntos
Apresentação de Antígeno , Células-Tronco Embrionárias/imunologia , Epigênese Genética , Antígenos de Histocompatibilidade , Células-Tronco Pluripotentes Induzidas/imunologia , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II , Humanos , Imunidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA