Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 11(16)2022 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-36010677

RESUMO

Spalt-Like Transcription Factor 4 (SALL4) is a critical factor for self-renewal ability and pluripotency of stem cells. On the other hand, various reports show tight relation of SALL4 to cancer occurrence and metastasis. SALL4 exerts its effects not only by inducing gene expression but also repressing a large cluster of genes through interaction with various epigenetic modifiers. Due to high expression of SALL4 in cancer cells and its silence in almost all adult tissues, it is an ideal target for cancer therapy. However, targeting SALL4 meets various challenges. SALL4 is a transcription factor and designing appropriate drug to inhibit this intra-nucleus component is challenging. On the other hand, due to lack of our knowledge on structure of the protein and the suitable active sites, it becomes more difficult to reach the appropriate drugs against SALL4. In this review, we have focused on approaches applied yet to target this oncogene and discuss the potential of degrader systems as new therapeutics to target oncogenes.


Assuntos
Neoplasias , Fatores de Transcrição , Adulto , Regulação da Expressão Gênica , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
2.
Nat Commun ; 13(1): 684, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115561

RESUMO

Loss of pancreatic beta cells is a central feature of type 1 (T1D) and type 2 (T2D) diabetes, but a therapeutic strategy to preserve beta cell mass remains to be established. Here we show that the death receptor TMEM219 is expressed on pancreatic beta cells and that signaling through its ligand insulin-like growth factor binding protein 3 (IGFBP3) leads to beta cell loss and dysfunction. Increased peripheral IGFBP3 was observed in established and at-risk T1D/T2D patients and was confirmed in T1D/T2D preclinical models, suggesting that dysfunctional IGFBP3/TMEM219 signaling is associated with abnormalities in beta cells homeostasis. In vitro and in vivo short-term IGFBP3/TMEM219 inhibition and TMEM219 genetic ablation preserved beta cells and prevented/delayed diabetes onset, while long-term IGFBP3/TMEM219 blockade allowed for beta cell expansion. Interestingly, in several patients' cohorts restoration of appropriate IGFBP3 levels was associated with improved beta cell function. The IGFBP3/TMEM219 pathway is thus shown to be a physiological regulator of beta cell homeostasis and is also demonstrated to be disrupted in T1D/T2D. IGFBP3/TMEM219 targeting may therefore serve as a therapeutic option in diabetes.


Assuntos
Regulação da Expressão Gênica , Homeostase/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/genética , Transdução de Sinais/genética , Adulto , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Humanos , Immunoblotting , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Int J Mol Sci ; 21(17)2020 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-32878275

RESUMO

Cell therapy with a variety of stem populations is increasingly being investigated as a promising regenerative strategy for cardiovascular (CV) diseases. Their combination with adequate scaffolds represents an improved therapeutic approach. Recently, several biomaterials were investigated as scaffolds for CV tissue repair, with decellularized extracellular matrices (dECMs) arousing increasing interest for cardiac tissue engineering applications. The aim of this study was to analyze whether dECMs support the cardiac differentiation of CardiopoieticAF stem cells. These perinatal stem cells, which can be easily isolated without ethical or safety limitations, display a high cardiac differentiative potential. Differentiation was previously achieved by culturing them on Matrigel, but this 3D scaffold is not transplantable. The identification of a new transplantable scaffold able to support CardiopoieticAF stem cell cardiac differentiation is pivotal prior to encouraging translation of in vitro studies in animal model preclinical investigations. Our data demonstrated that decellularized extracellular matrices already used in cardiac surgery (the porcine CorTMPATCH and the equine MatrixPatchTM) can efficiently support the proliferation and cardiac differentiation of CardiopoieticAF stem cells and represent a useful cellular scaffold to be transplanted with stem cells in animal hosts.


Assuntos
Líquido Amniótico/citologia , Diferenciação Celular , Matriz Extracelular/química , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Engenharia Tecidual , Alicerces Teciduais/química , Líquido Amniótico/metabolismo , Animais , Adesão Celular , Proliferação de Células , Colágeno , Combinação de Medicamentos , Matriz Extracelular/metabolismo , Feminino , Cavalos , Humanos , Laminina , Masculino , Miócitos Cardíacos/metabolismo , Proteoglicanas , Células-Tronco/metabolismo , Suínos
4.
Cells ; 9(5)2020 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-32456308

RESUMO

Human perinatal stem cells (SCs) can be isolated from fetal annexes without ethical or safety limitations. They are generally considered multipotent; nevertheless, their biological characteristics are still not fully understood. The aim of this study was to investigate the pluripotency potential of human perinatal SCs as compared to human induced pluripotent stem cells (hiPSCs). Despite the low expression of the pluripotent factors NANOG, OCT4, SOX2, and C-KIT in perinatal SC, we observed minor differences in the promoters DNA-methylation profile of these genes with respect to hiPSCs; we also demonstrated that in perinatal SCs miR-145-5p had an inverse trend in comparison to these stemness markers, suggesting that NANOG, OCT4, and SOX2 were regulated at the post-transcriptional level. The reduced expression of stemness markers was also associated with shorter telomere lengths and shift of the oxidative metabolism between hiPSCs and fetal annex-derived cells. Our findings indicate the differentiation ability of perinatal SCs might not be restricted to the mesenchymal lineage due to an epigenetic barrier, but other regulatory mechanisms such as telomere shortening or metabolic changes might impair their differentiation potential and challenge their clinical application.


Assuntos
Epigênese Genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Cromossomos Humanos/metabolismo , Metilação de DNA/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Recém-Nascido , MicroRNAs/genética , MicroRNAs/metabolismo , Regiões Promotoras Genéticas , Homeostase do Telômero
5.
Cells ; 7(1)2017 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-29280988

RESUMO

Red blood cells and platelets are anucleate blood components indispensable for oxygen delivery and hemostasis, respectively. Derivation of these blood elements from induced pluripotent stem (iPS) cells has the potential to develop blood donor-independent and genetic manipulation-prone products to complement or replace current transfusion banking, also minimizing the risk of alloimmunization. While the production of erythrocytes from iPS cells has challenges to overcome, such as differentiation into adult-type phenotype that functions properly after transfusion, platelet products are qualitatively and quantitatively approaching a clinically-applicable level owing to advances in expandable megakaryocyte (MK) lines, platelet-producing bioreactors, and novel reagents. Guidelines that assure the quality of iPS cells-derived blood products for clinical application represent a novel challenge for regulatory agencies. Considering the minimal risk of tumorigenicity and the expected significant demand of such products, ex vivo production of iPS-derived blood components can pave the way for iPS translation into the clinic.

6.
Mol Cell Oncol ; 3(3): e1093690, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27314082

RESUMO

Gene expression programs are tightly regulated by heritable "epigenetic" information, which is stored as chemical modifications of histones and DNA. With the recent development of sequencing-based epigenome mapping technologies and cancer cellular reprogramming, the tools are now in hand to analyze the epigenetic contribution to human cancer.

7.
Gene ; 584(2): 111-9, 2016 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26892498

RESUMO

There is a growing body of evidence supporting that cancer cells share many similarities with embryonic stem cells (ESCs). For example, aggressive cancers and ESCs share a common gene expression signature that includes hundreds of genes. Since ESC genes are not present in most adult tissues, they could be ideal candidate targets for cancer-specific diagnosis and treatment. This is an exciting cancer-targeting model. The major hurdle to test this model is to identify the key factors/pathway(s) within ESCs that are responsible for the cancer phenotype. SALL4 is one of few genes that can establish this link. The first publication of SALL4 is on its mutation in a human inherited disorder with multiple developmental defects. Since then, over 300 papers have been published on various aspects of this gene in stem cells, development, and cancers. This review aims to summarize our current knowledge of SALL4, including a SALL4-based approach to classify and target cancers. Many questions about this important gene still remain unanswered, specifically, on how this gene regulates cell fates at a molecular level. Understanding SALL4's molecular functions will allow development of specific targeted approaches in the future.


Assuntos
Neoplasias/genética , Células-Tronco/citologia , Fatores de Transcrição/genética , Humanos , Neoplasias/patologia
8.
Cancer Cell ; 27(5): 671-81, 2015 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-25965572

RESUMO

Leukemic cells disrupt normal patterns of blood cell formation, but little is understood about the mechanism. We investigated whether leukemic cells alter functions of normal hematopoietic stem and progenitor cells. Exposure to chronic myelogenous leukemia (CML) caused normal mouse hematopoietic progenitor cells to divide more readily, altered their differentiation, and reduced their reconstitution and self-renewal potential. Interestingly, the normal bystander cells acquired gene expression patterns resembling their malignant counterparts. Therefore, much of the leukemia signature is mediated by extrinsic factors. Indeed, IL-6 was responsible for most of these changes. Compatible results were obtained when human CML were cultured with normal human hematopoietic progenitor cells. Furthermore, neutralization of IL-6 prevented these changes and treated the disease.


Assuntos
Citocinas/antagonistas & inibidores , Células-Tronco Hematopoéticas/citologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Animais , Técnicas de Cocultura , Citocinas/metabolismo , Humanos , Interleucina-6/farmacologia , Interleucina-6/uso terapêutico , Camundongos , Células Tumorais Cultivadas
9.
Nat Commun ; 6: 7091, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25997600

RESUMO

Chronic myeloid leukaemia (CML) is a myeloproliferative disorder characterized by the genetic translocation t(9;22)(q34;q11.2) encoding for the BCR-ABL fusion oncogene. However, many molecular mechanisms of the disease progression still remain poorly understood. A growing body of evidence suggests that the epigenetic abnormalities are involved in tyrosine kinase resistance in CML, leading to leukaemic clone escape and disease propagation. Here we show that, by applying cellular reprogramming to primary CML cells, aberrant DNA methylation contributes to the disease evolution. Importantly, using a BCR-ABL inducible murine model, we demonstrate that a single oncogenic lesion triggers DNA methylation changes, which in turn act as a precipitating event in leukaemia progression.


Assuntos
Metilação de DNA , Genes abl , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Animais , Azacitidina , Diferenciação Celular , Técnicas de Reprogramação Celular , Humanos , Células K562 , Camundongos Transgênicos , Células U937
10.
Nat Chem Biol ; 10(8): 632-639, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24952596

RESUMO

The reprogramming of somatic cells to pluripotency using defined transcription factors holds great promise for biomedicine. However, human reprogramming remains inefficient and relies either on the use of the potentially dangerous oncogenes KLF4 and CMYC or the genetic inhibition of the tumor suppressor gene p53. We hypothesized that inhibition of signal transduction pathways that promote differentiation of the target somatic cells during development might relieve the requirement for non-core pluripotency factors during induced pluripotent stem cell (iPSC) reprogramming. Here, we show that inhibition of Notch greatly improves the efficiency of iPSC generation from mouse and human keratinocytes by suppressing p21 in a p53-independent manner and thereby enriching for undifferentiated cells capable of long-term self-renewal. Pharmacological inhibition of Notch enabled routine production of human iPSCs without KLF4 and CMYC while leaving p53 activity intact. Thus, restricting the development of somatic cells by altering intercellular communication enables the production of safer human iPSCs.


Assuntos
Oncogenes/fisiologia , Células-Tronco Pluripotentes/fisiologia , Receptores Notch/antagonistas & inibidores , Animais , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dipeptídeos/farmacologia , Genes myc , Genes p53 , Histona-Lisina N-Metiltransferase , Humanos , Queratinócitos/efeitos dos fármacos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Cancer Cell ; 24(5): 575-88, 2013 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-24183681

RESUMO

Mutation or epigenetic silencing of the transcription factor C/EBPα is observed in ∼10% of patients with acute myeloid leukemia (AML). In both cases, a common global gene expression profile is observed, but downstream targets relevant for leukemogenesis are not known. Here, we identify Sox4 as a direct target of C/EBPα whereby its expression is inversely correlated with C/EBPα activity. Downregulation of Sox4 abrogated increased self-renewal of leukemic cells and restored their differentiation. Gene expression profiles of leukemia-initiating cells (LICs) from both Sox4 overexpression and murine C/EBPα mutant AML models clustered together but differed from other types of AML. Our data demonstrate that Sox4 overexpression resulting from C/EBPα inactivation contributes to the development of leukemia with a distinct LIC phenotype.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Fatores de Transcrição SOXC/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Knockout , Mutação , Células Mieloides/metabolismo , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Oncogenes , Fatores de Transcrição SOXC/metabolismo , Transcriptoma
12.
Nature ; 503(7476): 371-6, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24107992

RESUMO

DNA methylation was first described almost a century ago; however, the rules governing its establishment and maintenance remain elusive. Here we present data demonstrating that active transcription regulates levels of genomic methylation. We identify a novel RNA arising from the CEBPA gene locus that is critical in regulating the local DNA methylation profile. This RNA binds to DNMT1 and prevents CEBPA gene locus methylation. Deep sequencing of transcripts associated with DNMT1 combined with genome-scale methylation and expression profiling extend the generality of this finding to numerous gene loci. Collectively, these results delineate the nature of DNMT1-RNA interactions and suggest strategies for gene-selective demethylation of therapeutic targets in human diseases.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Regulação da Expressão Gênica/genética , RNA não Traduzido/metabolismo , Sequência de Bases , Linhagem Celular , DNA/genética , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , Perfilação da Expressão Gênica , Genoma Humano/genética , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA não Traduzido/genética , Proteínas de Ligação a RNA/metabolismo , Especificidade por Substrato , Transcrição Gênica/genética
13.
Blood ; 121(20): 4073-81, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-23547051

RESUMO

Dendritic cells (DCs) are master regulators of the immune system, but molecular regulation of early DC differentiation has been poorly understood. Here, we report that the transcription factor C/EBPα coordinates the development of progenitor cells required for production of multiple categories of DCs. C/EBPα was needed for differentiation from stem/progenitor cells to common DC progenitors (CDPs), but not for transition of CDP to mature DCs. C/EBPα deletion in mature DCs did not affect their numbers or function, suggesting that this transcription factor is not needed for maintenance of DCs in lymphoid tissues. ChIP-seq and microarrays were used to identify candidate genes regulated by C/EBPα and required for DC formation. Genes previously shown to be critical for DC formation were bound by C/EBPα, and their expression was decreased in the earliest hematopoietic compartments in the absence of C/EBPα. These data indicate that C/EBPα is important for the earliest stages of steady-state DC differentiation.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/fisiologia , Diferenciação Celular/genética , Células Dendríticas/fisiologia , Células-Tronco/fisiologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Análise por Conglomerados , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco/metabolismo
14.
Nat Cell Biol ; 15(4): 385-94, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23502316

RESUMO

In blood, the transcription factor C/EBPa is essential for myeloid differentiation and has been implicated in regulating self-renewal of fetal liver haematopoietic stem cells (HSCs). However, its function in adult HSCs has remained unknown. Here, using an inducible knockout model we found that C/EBPa-deficient adult HSCs underwent a pronounced increase in number with enhanced proliferation, characteristics resembling fetal liver HSCs. Consistently, transcription profiling of C/EBPa-deficient HSCs revealed a gene expression program similar to fetal liver HSCs. Moreover, we observed that age-specific Cebpa expression correlated with its inhibitory effect on the HSC cell cycle. Mechanistically we identified N-Myc as a downstream target of C/EBPa, and loss of C/EBPa resulted in de-repression of N-Myc. Our data establish C/EBPa as a central determinant in the switch from fetal to adult HSCs.


Assuntos
Células-Tronco Adultas/citologia , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Diferenciação Celular , Proliferação de Células , Células-Tronco Fetais/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Adultas/fisiologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Transplante de Medula Óssea , Ciclo Celular , Imunoprecipitação da Cromatina , Células-Tronco Fetais/fisiologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Integrases/metabolismo , Luciferases/metabolismo , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Blood ; 121(8): 1255-64, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23212524

RESUMO

Lineage-restricted cells can be reprogrammed to a pluripotent state known as induced pluripotent stem (iPS) cells through overexpression of 4 transcription factors. iPS cells are similar to human embryonic stem (hES) cells and have the same ability to generate all the cells of the human body, including blood cells. However, this process is extremely inefficient and to date has been unsuccessful at differentiating iPS into hematopoietic stem cells (HSCs). We hypothesized that iPS cells, injected into NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ immunocompromised (NSG) mice could give rise to hematopoietic stem/progenitor cells (HSPCs) during teratoma formation. Here, we report a novel in vivo system in which human iPS cells differentiate within teratomas to derive functional myeloid and lymphoid cells. Similarly, HSPCs can be isolated from teratoma parenchyma and reconstitute a human immune system when transplanted into immunodeficient mice. Our data provide evidence that in vivo generation of patient customized cells is feasible, providing materials that could be useful for transplantation, human antibody generation, and drug screening applications.


Assuntos
Hematopoese/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Teratoma/patologia , Animais , Linfócitos B/citologia , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Queratinócitos/fisiologia , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células Mieloides/citologia , Transplante de Neoplasias , Células Estromais/citologia , Células Estromais/fisiologia , Células Estromais/transplante , Linfócitos T/citologia , Teratoma/genética , Transplante Heterólogo , Células Tumorais Cultivadas
16.
J Clin Invest ; 122(12): 4490-504, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23160200

RESUMO

C/EBPs are a family of transcription factors that regulate growth control and differentiation of various tissues. We found that C/EBPγ is highly upregulated in a subset of acute myeloid leukemia (AML) samples characterized by C/EBPα hypermethylation/silencing. Similarly, C/EBPγ was upregulated in murine hematopoietic stem/progenitor cells lacking C/EBPα, as C/EBPα mediates C/EBPγ suppression. Studies in myeloid cells demonstrated that CEBPG overexpression blocked neutrophilic differentiation. Further, downregulation of Cebpg in murine Cebpa-deficient stem/progenitor cells or in human CEBPA-silenced AML samples restored granulocytic differentiation. In addition, treatment of these leukemias with demethylating agents restored the C/EBPα-C/EBPγ balance and upregulated the expression of myeloid differentiation markers. Our results indicate that C/EBPγ mediates the myeloid differentiation arrest induced by C/EBPα deficiency and that targeting the C/EBPα-C/EBPγ axis rescues neutrophilic differentiation in this unique subset of AMLs.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Diferenciação Celular , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/metabolismo , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Metilação de DNA , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Epigênese Genética , Genes Reporter , Fator Estimulador de Colônias de Granulócitos/fisiologia , Granulócitos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Neutrófilos/metabolismo , Neutrófilos/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Regulação para Cima
17.
EMBO J ; 30(19): 4059-70, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21873977

RESUMO

The transcription factor RUNX1 is essential to establish the haematopoietic gene expression programme; however, the mechanism of how it activates transcription of haematopoietic stem cell (HSC) genes is still elusive. Here, we obtained novel insights into RUNX1 function by studying regulation of the human CD34 gene, which is expressed in HSCs. Using transgenic mice carrying human CD34 PAC constructs, we identified a novel downstream regulatory element (DRE), which is bound by RUNX1 and is necessary for human CD34 expression in long-term (LT)-HSCs. Conditional deletion of Runx1 in mice harbouring human CD34 promoter-DRE constructs abrogates human CD34 expression. We demonstrate by chromosome conformation capture assays in LT-HSCs that the DRE physically interacts with the human CD34 promoter. Targeted mutagenesis of RUNX binding sites leads to perturbation of this interaction and decreased human CD34 expression in LT-HSCs. Overall, our in vivo data provide novel evidence about the role of RUNX1 in mediating interactions between distal and proximal elements of the HSC gene CD34.


Assuntos
Antígenos CD34/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Animais , Transplante de Medula Óssea , Cromatina/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Sangue Fetal/citologia , Genótipo , Células HL-60 , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Sequências Reguladoras de Ácido Nucleico/genética
18.
Trends Mol Med ; 15(2): 59-68, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19162546

RESUMO

Lineage-restricted cells can be reprogrammed to a pluripotent state through overexpression of defined transcription factors. Here, we summarize recent progress in the direct reprogramming field and discuss data comparing embryonic stem (ES) and induced pluripotent stem (iPS) cells. Results from many independent groups suggest that mouse and human iPS cells, once established, generally exhibit a normal karyotype, are transcriptionally and epigenetically similar to ES cells and maintain the potential to differentiate into derivatives of all germ layers. Recent developments provide optimism that safe, viral-free human iPS cells could be derived routinely in the near future. An important next step will be to identify ways of assessing which iPS cell lines are sufficiently reprogrammed and safe to use for therapeutic applications. The approach of generating patient-specific pluripotent cells will undoubtedly transform regenerative medicine in many ways.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes/fisiologia , Medicina Regenerativa , Animais , Linhagem Celular , Epigênese Genética , Perfilação da Expressão Gênica , Genoma , Humanos
19.
Mol Cancer Ther ; 7(5): 1140-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18483302

RESUMO

The demonstration that the small synthetic molecule reversine [2-(4-morpholinoanilino)-N6-cyclohexyladenine] promotes the dedifferentiation of committed cells into multipotent progenitor-type cells has raised hopes on the exploitation of this small chemical tool for the generation of stem cells. Here, we show that reversine causes a failure in cytokinesis and induces polyploidization. These effects of reversine are due to the inhibition of Aurora A and B, two related kinases that are implicated in several aspects of mitosis and that are frequently amplified and overexpressed in human tumors. Reversine inhibits the phosphorylation of histone H3, a direct downstream target of Aurora kinases. Similarly to the Aurora kinase inhibitor VX-680, which has recently entered phase II clinical trials for cancer treatment, reversine inhibited colony formation of leukemic cells from patients with acute myeloid leukemia but was significantly less toxic than VX-680 on cells from healthy donors. The crystal structure of the reversine-Aurora B kinase complex shows that reversine is a novel class of ATP-competitive Aurora kinase inhibitors. Thus, although our studies raise serious doubts on the application of reversine in regenerative medicine, they support the paradigm that reversine might be a useful agent in cancer chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide Aguda/enzimologia , Morfolinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Purinas/farmacologia , Aurora Quinase B , Aurora Quinases , Sítios de Ligação/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Relação Dose-Resposta a Droga , Células HeLa , Histonas/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Modelos Moleculares , Morfolinas/química , Morfolinas/metabolismo , Fosforilação , Poliploidia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Purinas/química , Purinas/metabolismo
20.
Blood ; 109(4): 1460-71, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17038527

RESUMO

Recent evidence suggests that mutations in the Gata1 gene may alter the proliferation/differentiation potential of hemopoietic progenitors. By single-cell cloning and sequential replating experiments of prospectively isolated progenitor cells, we demonstrate here that the hypomorphic Gata1low mutation increases the proliferation potential of a unique class of progenitor cells, similar in phenotype to adult common erythroid/megakaryocytic progenitors (MEPs), but with the "unique" capacity to generate erythroblasts, megakaryocytes, and mast cells in vitro. Conversely, progenitor cells phenotypically similar to mast cell progenitors (MCPs) are not detectable in the marrow from these mutants. At the single-cell level, about 11% of Gata1low progenitor cells, including MEPs, generate cells that will continue to proliferate in cultures for up to 4 months. In agreement with these results, trilineage (erythroid, megakaryocytic, and mastocytic) cell lines are consistently isolated from bone marrow and spleen cells of Gata1low mice. These results confirm the crucial role played by Gata1 in hematopoietic commitment and identify, as a new target for the Gata1 action, the restriction point at which common myeloid progenitors become either MEPs or MCPs.


Assuntos
Diferenciação Celular/genética , Proliferação de Células , Células Precursoras Eritroides/citologia , Fator de Transcrição GATA1/genética , Megacariócitos/citologia , Mutação , Animais , Células da Medula Óssea , Células Cultivadas , Mastócitos/citologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...