Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 15(1): 193, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956724

RESUMO

BACKGROUND: The human induced pluripotent stem cells (hiPSCs) can generate all the cells composing the human body, theoretically. Therefore, hiPSCs are thought to be a candidate source of stem cells for regenerative medicine. The major challenge of allogeneic hiPSC-derived cell products is their immunogenicity. The hypoimmunogenic cell strategy is allogenic cell therapy without using immune suppressants. Advances in gene engineering technology now permit the generation of hypoimmunogenic cells to avoid allogeneic immune rejection. In this study, we generated a hypoimmunogenic hiPSC (HyPSC) clone that had diminished expression of human leukocyte antigen (HLA) class Ia and class II and expressed immune checkpoint molecules and a safety switch. METHODS: First, we generated HLA class Ia and class II double knockout (HLA class Ia/II DKO) hiPSCs. Then, a HyPSC clone was generated by introducing exogenous ß-2-microglobulin (B2M), HLA-G, PD-L1, and PD-L2 genes, and the Rapamycin-activated Caspase 9 (RapaCasp9)-based suicide gene as a safety switch into the HLA class Ia/II DKO hiPSCs. The characteristics and immunogenicity of the HyPSCs and their derivatives were analyzed. RESULTS: We found that the expression of HLA-G on the cell surface can be enhanced by introducing the exogenous HLA-G gene along with B2M gene into HLA class Ia/II DKO hiPSCs. The HyPSCs retained a normal karyotype and had the characteristics of pluripotent stem cells. Moreover, the HyPSCs could differentiate into cells of all three germ layer lineages including CD45+ hematopoietic progenitor cells (HPCs), functional endothelial cells, and hepatocytes. The HyPSCs-derived HPCs exhibited the ability to evade innate and adaptive immunity. Further, we demonstrated that RapaCasp9 could be used as a safety switch in vitro and in vivo. CONCLUSION: The HLA class Ia/II DKO hiPSCs armed with HLA-G, PD-L1, PD-L2, and RapaCasp9 molecules are a potential source of stem cells for allogeneic transplantation.


Assuntos
Imunidade Adaptativa , Antígeno B7-H1 , Antígenos HLA-G , Imunidade Inata , Células-Tronco Pluripotentes Induzidas , Proteína 2 Ligante de Morte Celular Programada 1 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/imunologia , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Antígenos HLA-G/genética , Antígenos HLA-G/metabolismo , Antígenos HLA-G/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Proteína 2 Ligante de Morte Celular Programada 1/genética , Animais , Camundongos
3.
Stem Cell Res Ther ; 14(1): 83, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37046329

RESUMO

Stem cell replacement holds the potential for sensorineural hearing loss (SNHL) treatment. However, its translation into clinical practice requires strategies for improving stem cell survival following intracochlear transplantation. Considering recent findings showing that the inner ear contains a resident population of immune cells, we hypothesized that immune evasion would improve the survival and residence time of transplanted stem cells in the cochlea, potentially leading to better outcomes. To test this, we leveraged genetic engineering techniques to develop hypoimmunogenic human-induced pluripotent stem cells (hi-iPSC), which lack human leukocyte antigen expression. We found that gene editing does not affect the biological properties of hi-iPSCs, including their capacity to differentiate into otic neural progenitors (ONPs). Compared to wild-type ONPs, more hypoimmunogenic ONPs (derived from hi-iPSCs) were found in the inner ear of immunocompetent mice ten days following cochlear xenotransplantation. This approach may open a new avenue for experimental and clinical SNHL treatments.


Assuntos
Perda Auditiva , Células-Tronco Pluripotentes Induzidas , Camundongos , Humanos , Animais , Transplante Heterólogo , Diferenciação Celular , Perda Auditiva/metabolismo , Transplante de Células-Tronco/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo
4.
Genes Dev ; 32(23-24): 1537-1549, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30463901

RESUMO

Human globin gene production transcriptionally "switches" from fetal to adult synthesis shortly after birth and is controlled by macromolecular complexes that enhance or suppress transcription by cis elements scattered throughout the locus. The DRED (direct repeat erythroid-definitive) repressor is recruited to the ε-globin and γ-globin promoters by the orphan nuclear receptors TR2 (NR2C1) and TR4 (NR2C2) to engender their silencing in adult erythroid cells. Here we found that nuclear receptor corepressor-1 (NCoR1) is a critical component of DRED that acts as a scaffold to unite the DNA-binding and epigenetic enzyme components (e.g., DNA methyltransferase 1 [DNMT1] and lysine-specific demethylase 1 [LSD1]) that elicit DRED function. We also describe a potent new regulator of γ-globin repression: The deubiquitinase BRCA1-associated protein-1 (BAP1) is a component of the repressor complex whose activity maintains NCoR1 at sites in the ß-globin locus, and BAP1 inhibition in erythroid cells massively induces γ-globin synthesis. These data provide new mechanistic insights through the discovery of novel epigenetic enzymes that mediate γ-globin gene repression.


Assuntos
Regulação da Expressão Gênica/genética , Correpressor 1 de Receptor Nuclear/genética , Correpressor 1 de Receptor Nuclear/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , gama-Globinas/genética , Sítios de Ligação , Linhagem Celular , Ativação Enzimática/genética , Epigênese Genética/genética , Células Eritroides/metabolismo , Inativação Gênica , Células HEK293 , Humanos , Células K562 , Membro 1 do Grupo C da Subfamília 2 de Receptores Nucleares/metabolismo , Domínios Proteicos , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo
5.
Sci Rep ; 8(1): 8037, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29795117

RESUMO

Numerous small molecules (termed inducers), many of which are electrophiles, upregulate cytoprotective responses and inhibit pro-inflammatory pathways by activating nuclear factor-erythroid 2 p45-related factor 2 (NRF2). Key to NRF2 activation is the ability to chemically modifying critical sensor cysteines in the main negative regulator of NRF2, Kelch-like ECH-associated protein 1 (KEAP1), of which C151, C273 and C288 are best characterized. This study aimed to establish the requirement for these cysteine sensor(s) for the biological activities of the most potent NRF2 activators known to date, the cyclic cyanoenones, some of which are in clinical trials. It was found that C151 in KEAP1 is the main cysteine sensor for this class of inducers, irrespective of molecular size or shape. Furthermore, in primary macrophage cells expressing C151S mutant KEAP1, at low concentrations, the tricyclic cyanoenone TBE-31 is inactive as an activator of NRF2 as well as an inhibitor of lipopolysaccharide-stimulated gene expression of the pro-inflammatory cytokines IL6 and IL1ß. However, at high inducer concentrations, NRF2 activation proceeds in the absence of C151, albeit at a lower magnitude. Our findings highlight the intrinsic flexibility of KEAP1 and emphasize the critical importance of establishing the precise dose of NRF2 activators for maintaining on-target selectivity.


Assuntos
Cisteína/química , Proteína 1 Associada a ECH Semelhante a Kelch/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Fenantrenos/farmacologia , Ativação Transcricional/efeitos dos fármacos , Animais , Células Cultivadas , Cisteína/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/genética , Fenantrenos/química , Regulação para Cima
6.
Genes Dev ; 29(18): 1930-41, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26385963

RESUMO

Protein abundance must be precisely regulated throughout life, and nowhere is the stringency of this requirement more evident than during T-cell development: A twofold increase in the abundance of transcription factor GATA3 results in thymic lymphoma, while reduced GATA3 leads to diminished T-cell production. GATA3 haploinsufficiency also causes human HDR (hypoparathyroidism, deafness, and renal dysplasia) syndrome, often accompanied by immunodeficiency. Here we show that loss of one Gata3 allele leads to diminished expansion (and compromised development) of immature T cells as well as aberrant induction of myeloid transcription factor PU.1. This effect is at least in part mediated transcriptionally: We discovered that Gata3 is monoallelically expressed in a parent of origin-independent manner in hematopoietic stem cells and early T-cell progenitors. Curiously, half of the developing cells switch to biallelic Gata3 transcription abruptly at midthymopoiesis. We show that the monoallelic-to-biallelic transcriptional switch is stably maintained and therefore is not a stochastic phenomenon. This unique mechanism, if adopted by other regulatory genes, may provide new biological insights into the rather prevalent phenomenon of monoallelic expression of autosomal genes as well as into the variably penetrant pathophysiological spectrum of phenotypes observed in many human syndromes that are due to haploinsufficiency of the affected gene.


Assuntos
Alelos , Fator de Transcrição GATA3/genética , Regulação da Expressão Gênica/genética , Linfócitos T/metabolismo , Animais , Medula Óssea/metabolismo , Proliferação de Células/genética , Células Cultivadas , Fator de Transcrição GATA3/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/genética , Timócitos/citologia , Timócitos/metabolismo , Transativadores/genética
7.
Blood ; 122(23): 3798-807, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24092935

RESUMO

In previous mass spectrometry and coimmune precipitation studies, we identified tripartite motif-containing 28 (TRIM28; also known as transcriptional intermediary factor1ß and Krüppel-associated box-associated protein-1) as a cofactor that specifically copurified with an NR2C1/NR2C2 (TR2/TR4) orphan nuclear receptor heterodimer that previous studies had implicated as an embryonic/fetal ß-type globin gene repressor. TRIM28 has been characterized as a transcriptional corepressor that can associate with many different transcription factors and can play functional roles in multiple tissues and cell types. Here, we tested the contribution of TRIM28 to globin gene regulation and erythropoiesis using a conditional loss-of-function in vivo model. We discovered that Trim28 genetic loss in the adult mouse leads to defective immature erythropoiesis in the bone marrow and consequently to anemia. We further found that TRIM28 controls erythropoiesis in a cell-autonomous manner by inducibly deleting Trim28 exclusively in hematopoietic cells. Finally, in the absence of TRIM28, we observed increased apoptosis as well as diminished expression of multiple erythroid transcription factors and heme biosynthetic enzymes in immature erythroid cells. Thus, TRIM28 is essential for the cell-autonomous development of immature erythroblasts in the bone marrow.


Assuntos
Eritroblastos/citologia , Eritroblastos/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Anemia/genética , Anemia/patologia , Animais , Apoptose/genética , Apoptose/fisiologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Eritropoese/genética , Eritropoese/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido , Globinas beta/genética
8.
Free Radic Biol Med ; 53(12): 2256-62, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23051009

RESUMO

Atherosclerosis is the major etiology underlying myocardial infarction and stroke, and strategies for preventing atherosclerosis are urgently needed. In the context of atherosclerosis, the deletion of the Nrf2 gene, which encodes a master regulator of the oxidative stress response in mammals, reportedly attenuates atherosclerosis formation. However, the precise mechanisms of protection against atherosclerosis are largely unknown. To further clarify the role of Nrf2 in atherosclerosis in vivo, we performed a time course analysis of atherosclerosis development utilizing an ApoE knockout (KO) mouse model. The results demonstrate that oil red O-stainable lesions were similar in size 5 weeks after the initiation of an HFC (high fat and high cholesterol) diet, but the lesions were markedly attenuated in the Nrf2 and ApoE double KO mice (A0N0 mice) compared with the lesions in the ApoE KO mice (A0N2 mice) at 12 weeks. Consistent with these results, the immunohistochemical analysis revealed that Nrf2 activation is observed in late-stage atherosclerotic plaques but not in earlier lesions. The RT-qPCR analysis of 12-week atherosclerotic plaques revealed that Nrf2 target genes, such as Ho-1 and SLPI, are expressed at significantly lower levels in the A0N0 mice compared with the A0N2 mice, and this change was associated with a decreased expression of macrophage M1-subtype genes Arginase II and inducible NO synthase in the A0N0 mice. Furthermore, the bone marrow (BM) transplantation (BMT) analysis revealed that the Nrf2 activity in the BM-derived cells contributed to lesion formation. Therefore, our study has characterized the positive role of Nrf2 in the BM-derived cells during the development of atherosclerosis, which suggests that Nrf2 may influence the inflammatory reactions in the plaques.


Assuntos
Aterosclerose/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Placa Aterosclerótica/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/imunologia , Aterosclerose/patologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Feminino , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Ativação Transcricional
9.
J Clin Invest ; 122(10): 3705-17, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22996665

RESUMO

The transcription factor GATA-2 plays vital roles in quite diverse developmental programs, including hematopoietic stem cell (HSC) survival and proliferation. We previously identified a vascular endothelial (VE) enhancer that regulates GATA-2 activity in pan-endothelial cells. To more thoroughly define the in vivo regulatory properties of this enhancer, we generated a tamoxifen-inducible Cre transgenic mouse line using the Gata2 VE enhancer (Gata2 VECre) and utilized it to temporally direct tissue-specific conditional loss of Gata2. Here, we report that Gata2 VECre-mediated loss of GATA-2 led to anemia, hemorrhage, and eventual death in edematous embryos. We further determined that the etiology of anemia in conditional Gata2 mutant embryos involved HSC loss in the fetal liver, as demonstrated by in vitro colony-forming and immunophenotypic as well as in vivo long-term competitive repopulation experiments. We further documented that the edema and hemorrhage in conditional Gata2 mutant embryos were due to defective lymphatic development. Thus, we unexpectedly discovered that in addition to its contribution to endothelial cell development, the VE enhancer also regulates GATA-2 expression in definitive fetal liver and adult BM HSCs, and that GATA-2 function is required for proper lymphatic vascular development during embryogenesis.


Assuntos
Anemia/genética , Elementos Facilitadores Genéticos , Morte Fetal/genética , Fator de Transcrição GATA2/fisiologia , Hematopoese/genética , Hemorragia/genética , Sistema Linfático/embriologia , Anemia/embriologia , Animais , Divisão Celular , Sobrevivência Celular , Ensaio de Unidades Formadoras de Colônias , Feminino , Fator de Transcrição GATA2/deficiência , Fator de Transcrição GATA2/genética , Genes Reporter , Células-Tronco Hematopoéticas/patologia , Hemorragia/embriologia , Imunofenotipagem , Fígado/citologia , Fígado/embriologia , Sistema Linfático/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Gravidez , Tamoxifeno/farmacologia
10.
Blood ; 119(10): 2242-51, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-22267605

RESUMO

Maintaining hematopoietic stem cell (HSC) quiescence is a critical property for the life-long generation of blood cells. Approximately 75% of cells in a highly enriched long-term repopulating HSC (LT-HSC) pool (Lin(-)Sca1(+)c-Kit(hi)CD150(+)CD48(-)) are quiescent, with only a small percentage of the LT-HSCs in cycle. Transcription factor GATA-3 is known to be vital for the development of T cells at multiple stages in the thymus and for Th2 differentiation in the peripheral organs. Although it is well documented that GATA-3 is expressed in HSCs, a role for GATA-3 in any prethymic progenitor cell has not been established. In the present study, we show that Gata3-null mutant mice generate fewer LT-HSCs and that fewer Gata3-null LT-HSCs are in cycle. Furthermore, Gata3 mutant hematopoietic progenitor cells fail to be recruited into an increased cycling state after 5-fluorouracil-induced myelosuppression. Therefore, GATA-3 is required for the maintenance of a normal number of LT-HSCs and for their entry into the cell cycle.


Assuntos
Ciclo Celular/genética , Fator de Transcrição GATA3/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Proliferação de Células , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Hematopoese/genética , Transplante de Células-Tronco Hematopoéticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Arch Biochem Biophys ; 508(1): 101-9, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21303654

RESUMO

Iron is an essential element of hemoglobin, and efficient iron recycling from senescent erythrocytes by splenic macrophages is required for erythrocyte hemoglobin synthesis during erythropoiesis. Ferroportin 1 (Fpn1) is the sole iron exporter in mammals, and it also regulates iron reutilization. In this study, we demonstrated genetically that a redox-sensitive transcription factor, Nrf2, regulates Fpn1 mRNA expression in macrophages. Nrf2 activation by several electrophilic compounds commonly resulted in the upregulation of Fpn1 mRNA in bone marrow-derived and peritoneal macrophages obtained from wild-type mice but not from Nrf2 knockout mice. Further, Nrf2 activation enhanced iron release from the J774.1 murine macrophage cell line. Previous studies showed that inflammatory stimuli, such as LPS, downregulates macrophage Fpn1 by transcriptional and hepcidin-mediated post-translational mechanisms leading to iron sequestration by macrophages. We showed that two Nrf2 activators, diethyl maleate and sulforaphane (SFN; a natural Nrf2 activator found in broccoli), restored the LPS-induced suppression of Fpn1 mRNA in human and mouse macrophages, respectively. Furthermore, SFN counteracted the LPS-induced increase of Hepcidin mRNA by an Nrf2-independent mechanism in mouse peritoneal macrophages. These results demonstrate that Nrf2 regulates iron efflux from macrophages through Fpn1 gene transcription and suggest that Nrf2 may control iron metabolism during inflammation.


Assuntos
Proteínas de Transporte de Cátions/genética , Regulação para Baixo/efeitos dos fármacos , Ferro/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Indução Enzimática/efeitos dos fármacos , Heme Oxigenase-1/biossíntese , Hepcidinas , Humanos , Inflamação/genética , Inflamação/metabolismo , Isotiocianatos , Maleatos/farmacologia , Camundongos , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sulfóxidos , Tiocianatos/farmacologia , Regulação para Cima/efeitos dos fármacos
12.
Genes Cells ; 15(1): 77-89, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20015225

RESUMO

The first step of heme biosynthesis in animals is catalyzed by 5-aminolevulinate synthase (ALAS), which controls heme supply in various tissues. To clarify the roles that the nonspecific isoform of ALAS (ALAS-N) plays in vivo, we prepared a green fluorescent protein (GFP) knock-in mouse line in which the Alas1 gene (encoding ALAS-N) is replaced with a gfp gene. We found that mice bearing a homozygous knock-in allele (Alas1(GFP/GFP)) were lethal by embryonic day 8.5, demonstrating that ALAS-N is essential for early embryogenesis. Fluorescence microscopic and flow cytometric analyses of heterozygous mouse (Alas1(+/GFP)) tissues showed that the Alas1 expression level differs substantially in tissues; Alas1 is highly expressed in testis Leydig cells, exocrine glands (including submandibular and parotid glands), endocrine glands (such as adrenal and thyroid glands) and hematopoietic lineage cells (including neutrophils and eosinophils). Quantitative analyses of GFP mRNA and ALAS-N mRNA in various tissues of Alas1(+/GFP) mice suggested that the destabilization of ALAS-N mRNA was not uniform in the various tissues. These results thus lay bare that elaborate control of the endogenous heme supply operates in various mouse tissues through regulation of the ALAS-N expression level and that this control is essential for heme homeostasis in animals.


Assuntos
5-Aminolevulinato Sintetase/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , 5-Aminolevulinato Sintetase/metabolismo , Envelhecimento/genética , Animais , Linhagem da Célula/genética , Ritmo Circadiano/genética , Perda do Embrião/enzimologia , Embrião de Mamíferos/enzimologia , Embrião de Mamíferos/patologia , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Sistema Hematopoético/citologia , Sistema Hematopoético/metabolismo , Heterozigoto , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Especificidade de Órgãos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
J Exp Med ; 206(13): 2987-3000, 2009 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-19934022

RESUMO

Most T lymphocytes appear to arise from very rare early T lineage progenitors (ETPs) in the thymus, but the transcriptional programs that specify ETP generation are not completely known. The transcription factor GATA-3 is required for the development of T lymphocytes at multiple late differentiation steps as well as for the development of thymic natural killer cells. However, a role for GATA-3 before the double-negative (DN) 3 stage of T cell development has to date been obscured both by the developmental heterogeneity of DN1 thymocytes and the paucity of ETPs. We provide multiple lines of in vivo evidence through the analysis of T cell development in Gata3 hypomorphic mutant embryos, in irradiated mice reconstituted with Gata3 mutant hematopoietic cells, and in mice conditionally ablated for the Gata3 gene to show that GATA-3 is required for ETP generation. We further show that Gata3 loss does not affect hematopoietic stem cells or multipotent hematopoietic progenitors. Finally, we demonstrate that Gata3 mutant lymphoid progenitors exhibit neither increased apoptosis nor diminished cell-cycle progression. Thus, GATA-3 is required for the cell-autonomous development of the earliest characterized thymic T cell progenitors.


Assuntos
Linhagem da Célula , Fator de Transcrição GATA3/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Linfopoese , Linfócitos T/fisiologia , Animais , Fígado/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Timo/embriologia
14.
Biochem Biophys Res Commun ; 365(3): 562-7, 2008 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-18022386

RESUMO

AhR repressor (AhRR) is an AhR-related bHLH-PAS transcription factor. It is known to repress AhR transcription activity in a competitive manner. To examine AhRR functions in mice, we produced AhRR-deficient mice by gene knockout. AhRR(-/-) mice were born in normal Mendelian proportions, grew well, and were fertile. AhR(-/-) mice exhibited higher levels of Cyp1a1 (Cytochrome P450 1A1) mRNA induction in the skin, stomach and spleen than wild-type mice, while expression of Cyp1a1 mRNA was not significantly altered in the liver, lung, heart or other tissues, suggesting that "super-induction" of Cyp1a1 mRNA expression in AhRR(-/-) mice occurs in a tissue specific manner. AhRR(-/-) mice displayed a delayed response to skin carcinogenesis caused by benzo[a]pyrene. Since CYP1A1 is involved in the metabolic activation and detoxification of chemical carcinogens, these results suggest that overexpression of CYP1A1 shifts the balance of the metabolic activities in the skin of AhRR(-/-) mice in favor of the detoxification of carcinogens.


Assuntos
Benzo(a)pireno/toxicidade , Transformação Celular Neoplásica/genética , Citocromo P-450 CYP1A1/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Repressoras/fisiologia , Neoplasias Cutâneas/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Citocromo P-450 CYP1A1/análise , Citocromo P-450 CYP1A1/metabolismo , Expressão Gênica , Marcação de Genes , Inativação Metabólica/genética , Camundongos , Camundongos Mutantes , Proteínas Repressoras/análise , Proteínas Repressoras/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Distribuição Tecidual
15.
Biochem J ; 404(3): 459-66, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17313370

RESUMO

The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) contains two transcription activation domains, Neh4 (Nrf2 ECH homology 4) and Neh5, which co-ordinately regulate transactivation of cytoprotective genes. In the present study we aimed to clarify the role of the Neh5 domain in Nrf2-mediated gene regulation. Deletion of the complete Neh5 domain reduces expression of endogenous Nrf2 target genes, such as HO-1 (haem oxygenase 1), NQO1 [NAD(P)H:quinone oxidoreductase 1] and GCLM (glutamate cysteine ligase modulatory subunit), in human kidney epithelial cells. Furthermore, the deletion of Neh5 markedly repressed CBP [CREB (cAMP-response-element-binding protein)-binding protein] and BRG1 (Brahma-related gene 1) from associating with Nrf2, diminishing their co-operative enhancement of HO-1 promoter activity. Mutational analysis of the Neh5 domain revealed a motif that shares significant homology with beta-actin and ARP1 (actin-related protein 1). Mutagenesis of this motif selectively decreased HO-1, but not NQO1 and GCLM, expression. Taken together, these results indicate that the Neh5 domain has the ability to regulate Nrf2 target gene transcription, yet the role of the Neh5 domain in transcription varies from gene to gene.


Assuntos
Citoproteção/genética , Fator 2 Relacionado a NF-E2/química , Fator 2 Relacionado a NF-E2/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Animais , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Linhagem Celular , DNA Helicases/genética , DNA Helicases/metabolismo , Genes Reporter , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Fator 2 Relacionado a NF-E2/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
J Immunol ; 175(10): 6968-75, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16272357

RESUMO

Emphysema is one of the major pathological abnormalities associated with chronic obstructive pulmonary disease. The protease/antiprotease imbalance and inflammation resulting from oxidative stress have been attributed to the pathogenesis of emphysema. Nrf2 is believed to protect against oxidative tissue damage through the transcriptional activation of a battery of antioxidant enzymes. In this study, we investigated the protective role of Nrf2 in the development of emphysema using elastase-induced emphysema as our model system. We found that elastase-provoked emphysema was markedly exacerbated in Nrf2-knockout (KO) mice compared with wild-type mice. The severity of emphysema in Nrf2-KO mice correlated intimately with the degree of lung inflammation in the initial stage of elastase treatment. The highly inducible expression of antioxidant and antiprotease genes observed in wild-type alveolar macrophages was significantly attenuated in the lungs of Nrf2-KO mice. Interestingly, transplantation of wild-type bone marrow cells into Nrf2-KO mice retarded the development of initial lung inflammation and subsequent emphysema, and this improvement correlated well with the appearance of macrophages expressing Nrf2-regulated antiprotease and antioxidant genes. Thus, Nrf2 appears to exert its protective effects through the transcriptional activation of antiprotease and antioxidant genes in alveolar macrophages.


Assuntos
Enfisema/prevenção & controle , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Antioxidantes/metabolismo , Transplante de Medula Óssea , Enfisema/etiologia , Enfisema/metabolismo , Enfisema/patologia , Expressão Gênica , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/prevenção & controle , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Elastase Pancreática/toxicidade , Pneumonia/etiologia , Pneumonia/metabolismo , Pneumonia/patologia , Pneumonia/prevenção & controle , Inibidores de Proteases/metabolismo
17.
J Biol Chem ; 280(29): 27244-50, 2005 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-15917255

RESUMO

The Nrf2-Keap1 system coordinately regulates cytoprotective gene expression via the antioxidant responsive element (ARE). The expression of several ARE-regulated genes was found to be up-regulated in endothelial cells by laminar shear stress, suggesting that Nrf2 contributes to the anti-atherosclerosis response via the ARE. To gain further insight into the roles that Nrf2 plays in the development of atherosclerosis, we examined how Nrf2 regulates gene expression in response to anti-atherogenic laminar flow (L-flow) or pro-atherogenic oscillatory flow (O-flow). Exposure of human aortic endothelial cells (HAECs) to L-flow, but not to O-flow, induced the expression of cytoprotective genes, such as NAD(P)H quinone oxidoreductase 1 (NQO1) by 5-fold and heme oxygenase-1 by 8-fold. The critical contribution of Nrf2 to the expression induced by L-flow was ascertained in siRNA-mediated knock-down experiments. Two cyclooxygenase-2 (COX-2) specific inhibitors attenuated Nrf2 nuclear accumulation in the acute phase of L-flow exposure. A downstream product of COX-2, 15-deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2), activated the Nrf2 regulatory pathway in HAECs through binding to the cysteines of Keap1. These results demonstrate that 15d-PGJ2 is essential for L-flow to activate Nrf2 and induce anti-atherosclerotic gene expression. Whereas both L-flow and O-flow induced the nuclear accumulation of Nrf2 to comparable levels, chromatin immunoprecipitation analysis revealed that Nrf2 binding to the NQO1 ARE was significantly diminished in the case of O-flow compared with that of L-flow. These results suggest that O-flow inhibits Nrf2 activity at the DNA binding step, thereby suppressing athero-protective gene expression and hence predisposing the blood vessels to the formation of atherosclerosis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Endotélio Vascular/citologia , Proteínas/fisiologia , Transativadores/fisiologia , Aorta , Arteriosclerose/etiologia , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteína 1 Associada a ECH Semelhante a Kelch , NAD(P)H Desidrogenase (Quinona)/genética , Fator 2 Relacionado a NF-E2 , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Prostaglandina D2/fisiologia , Proteínas/metabolismo , RNA Interferente Pequeno , Elementos de Resposta , Estresse Mecânico , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA