Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cells ; 11(8)2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35455969

RESUMO

Vascular endothelial growth factor A (VEGF-A) is a secreted protein that stimulates angiogenesis in response to hypoxia. Under hypoxic conditions, a non-canonical long isoform called L-VEGF is concomitantly expressed with VEGF-A. Once translated, L-VEGF is proteolytically cleaved to generate N-VEGF and VEGF-A. Interestingly, while VEGF-A is secreted and affects the surrounding cells, N-VEGF is mobilized to the nucleus. This suggests that N-VEGF participates in transcriptional response to hypoxia. In this study, we performed a series of complementary experiments to examine the functional role of N-VEGF. Strikingly, we found that the mere expression of N-VEGF followed by its hypoxia-independent mobilization to the nucleus was sufficient to induce key genes associated with angiogenesis, such as Hif1α,VEGF-A isoforms, as well as genes associated with cell survival under hypoxia. Complementarily, when N-VEGF was genetically depleted, key hypoxia-induced genes were downregulated and cells were significantly susceptible to hypoxia-mediated apoptosis. This is the first report of N-VEGF serving as an autoregulatory arm of VEGF-A. Further experiments will be needed to determine the role of N-VEGF in cancer and embryogenesis.


Assuntos
Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular , Apoptose , Hipóxia Celular , Humanos , Hipóxia , Neovascularização Patológica/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
J Mol Biol ; 432(16): 4544-4560, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32534063

RESUMO

The regulation of gene expression is a result of a complex interplay between chromatin remodeling, transcription factors, and signaling molecules. Cell differentiation is accompanied by chromatin remodeling of specific loci to permanently silence genes that are not essential for the differentiated cell activity. The molecular cues that recruit the chromatin remodeling machinery are not well characterized. IRF8 is an immune-cell specific transcription factor and its expression is augmented by interferon-γ. Therefore, it serves as a model gene to elucidate the molecular mechanisms governing its silencing in non-immune cells. Ahigh-throughput shRNA library screen in IRF8 expression-restrictive cells enabled the identification of MafK as modulator of IRF8 silencing, affecting chromatin architecture. ChIP-Seq analysis revealed three MafK binding regions (-25 kb, -20 kb, and IRF8 6th intron) within the IRF8 locus. These MafK binding sites are sufficient to repress a reporter gene when cloned in genome-integrated lentiviral reporter constructs in only expression-restrictive cells. Conversely, plasmid-based constructs do not demonstrate such repressive effect. These results highlight the role of these MafK binding sites in mediating repressed chromatin assembly. Finally, a more thorough genomic analysis was performed, using CRISPR-Cas9 to delete MafK-int6 binding region in IRF8 expression-restrictive cells. Deleted clones exhibited an accessible chromatin conformation within the IRF8 locus that was accompanied by a significant increase in basal expression of IRF8 that was further induced by interferon-γ. Taken together, we identified and characterized several MafK binding elements within the IRF8 locus that mediate repressive chromatin conformation resulting in the silencing of IRF8 expression in a celltype-specific manner.


Assuntos
Cromatina/metabolismo , Fatores Reguladores de Interferon/genética , Fator de Transcrição MafK/genética , Fator de Transcrição MafK/metabolismo , Animais , Sítios de Ligação , Sistemas CRISPR-Cas , Montagem e Desmontagem da Cromatina , Regulação da Expressão Gênica , Células HEK293 , Humanos , Fatores Reguladores de Interferon/química , Fatores Reguladores de Interferon/metabolismo , Camundongos , Células NIH 3T3 , Especificidade de Órgãos , Células RAW 264.7 , RNA Interferente Pequeno/farmacologia
3.
J Mol Biol ; 431(2): 210-222, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30502383

RESUMO

Interferon regulatory factor 8 (IRF8) is a nuclear transcription factor that plays a key role in the hierarchical differentiation of hematopoietic stem cells toward monocyte/dendritic cell lineages. Therefore, its expression is mainly limited to bone marrow-derived cells. The molecular mechanisms governing this cell-type-restricted expression have been described. However, the molecular mechanisms that are responsible for its silencing in non-hematopoietic cells are elusive. Recently, we demonstrated a role for IRF8 third intron in restricting its expression in non-hematopoietic cells. Furthermore, we showed that this intron alone is sufficient to promote repressed chromatin a cell-type-specific manner. Here we demonstrate the effect of the IRF8 third intron on chromatin conformation during murine embryonal stem cell differentiation. Using genome editing, we provide data showing that the third intron plays a key role in priming the chromatin state of the IRF8 locus during cell differentiation. It mediates dual regulatory effects in a cell-type-specific mode. It acts as a repressor element governing chromatin state of the IRF8 locus during embryonal stem cell differentiation to cardiomyocytes that are expression-restrictive cells. Conversely, it functions as an activator element that is essential for open chromatin structure during the differentiation of these cells to dendritic cells that are expression-permissive cells. Together, these results point to the role of IRF8 third intron as a cell-type-specific chromatin priming element during embryonal stem cell differentiation. These data add another layer to our understanding of the molecular mechanisms governing misexpression of a cell-type-specific gene such as IRF8.


Assuntos
Diferenciação Celular/genética , Cromatina/genética , Fatores Reguladores de Interferon/genética , Íntrons/genética , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Linhagem Celular , Linhagem da Célula/genética , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/genética , Células HEK293 , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Monócitos/metabolismo , Miócitos Cardíacos/metabolismo , Células NIH 3T3 , Células RAW 264.7
4.
Mol Immunol ; 44(4): 338-46, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16597464

RESUMO

Interferon regulatory factor 1 (IRF-1) and IRF-8, also known as interferon consensus sequence binding protein (ICSBP), are important regulators of macrophage differentiation and function. These factors exert their activities through the formation of heterocomplexes. As such, they are coactivators of various interferon-inducible genes in macrophages. To gain better insights into the involvement of these two transcription factors in the onset of the innate immune response and to identify their regulatory network in activated macrophages, DNA microarray was employed. Changes in the expression profile were analyzed in peritoneal macrophages from wild type mice and compared to IRF-1 and IRF-8 null mice, before and following 4 h exposure to IFN-gamma and LPS. The expression pattern of 265 genes was significantly changed (up/down) in peritoneal macrophages extracted from wild type mice following treatment with IFN-gamma and LPS, while no changes in the expression levels of these genes were observed in samples of the same cell-type from both IRF-1 and IRF-8 null mice. Among these putative target genes, numerous genes are involved in macrophage activity during inflammation. The expression profile of 10 of them was further examined by quantitative RT-PCR. In addition, the promoter regions of three of the identified genes were analyzed by reporter gene assay for the ability to respond to IRF-1 and IRF-8. Together, our results suggest that both IRF-1 and IRF-8 are involved in the transcriptional regulation of these genes. We therefore suggest a broader role for IRF-1 and IRF-8 in macrophages differentiation and maturation, being important inflammatory mediators.


Assuntos
Fator Regulador 1 de Interferon/genética , Fatores Reguladores de Interferon/genética , Ativação de Macrófagos/genética , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Fator Regulador 1 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Ativação Transcricional/efeitos dos fármacos
5.
PLoS One ; 11(6): e0156812, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27257682

RESUMO

Interferon Regulatory Factor-8 (IRF-8) serves as a key factor in the hierarchical differentiation towards monocyte/dendritic cell lineages. While much insight has been accumulated into the mechanisms essential for its hematopoietic specific expression, the mode of restricting IRF-8 expression in non-hematopoietic cells is still unknown. Here we show that the repression of IRF-8 expression in restrictive cells is mediated by its 3rd intron. Removal of this intron alleviates the repression of Bacterial Artificial Chromosome (BAC) IRF-8 reporter gene in these cells. Fine deletion analysis points to conserved regions within this intron mediating its restricted expression. Further, the intron alone selectively initiates gene silencing only in expression-restrictive cells. Characterization of this intron's properties points to its role as an initiator of sustainable gene silencing inducing chromatin condensation with suppressive histone modifications. This intronic element cannot silence episomal transgene expression underlining a strict chromatin-dependent silencing mechanism. We validated this chromatin-state specificity of IRF-8 intron upon in-vitro differentiation of induced pluripotent stem cells (iPSCs) into cardiomyocytes. Taken together, the IRF-8 3rd intron is sufficient and necessary to initiate gene silencing in non-hematopoietic cells, highlighting its role as a nucleation core for repressed chromatin during differentiation.


Assuntos
Cromatina/metabolismo , Fatores Reguladores de Interferon/metabolismo , Íntrons/genética , Macrófagos/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Citometria de Fluxo , Humanos , Fatores Reguladores de Interferon/genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Células NIH 3T3 , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real
6.
J Interferon Cytokine Res ; 22(1): 153-60, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11846986

RESUMO

Interferon (IFN) consensus sequence binding protein (ICSBP) is a member of a family of transcription factors termed IFN regulatory factors (IRF) and is also called IRF-8. Its expression is restricted mainly to cells of the immune system, and it plays a key role in the maturation of macrophages. ICSBP exerts its activity through the formation of different DNA-binding heterocomplexes. The interacting partner dictates a specific DNA recognition sequence, thus rendering ICSBP dual transcriptional activity, that is, repression or activation. Accordingly, such DNA elements were identified at the promoter regions of target genes that manifest macrophage action. A specific module (IRF association domain [IAD]) within ICSBP and a PEST domain located on the interacting partners mediate this association. Thus, ICSBP serves as an excellent prototype, demonstrating how a small subset of transcription factors can regulate gene expression in a spatial, temporal, and delicate tuning through combinatorial protein-protein interactions on different enhanceasomes.


Assuntos
Proteínas Repressoras/metabolismo , Ativação Transcricional , Animais , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Fatores Reguladores de Interferon , Substâncias Macromoleculares , Macrófagos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Alinhamento de Sequência , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Int J Food Microbiol ; 74(3): 217-27, 2002 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-11981972

RESUMO

Felix 01 (F01) is a bacteriophage originally isolated by Felix and Callow which lyses almost all Salmonella strains and has been widely used as a diagnostic test for this genus. Molecular information about this phage is entirely lacking. In the present study, the DNA of the phage was found to be a double-stranded linear molecule of about 80 kb. 11.5 kb has been sequenced and in this region A + T content is 60%. There are relatively few restriction endonuclease cleavage sites in the native genome and clones show this is due to their absence rather than modification. A restriction map of the genome has been constructed. The ends of the molecule cannot be ligated although they contain 5' phosphates. At least 60% of the genome must encode proteins. In the sequenced portion, many open reading frames exist and these are tightly packed together. These have been examined for homology to published proteins but only 1 to 17 shows similarity to known proteins. F01 is therefore the prototype of a new phage family. On the basis of restriction sites, codon usage and the distribution of nonsense codons in the unused reading frames, a strong case can be made for natural selection that reacts to mRNA structure and function.


Assuntos
DNA Viral , Fagos de Salmonella/genética , Sequência de Bases , Códon , DNA Viral/química , DNA Viral/genética , DNA Viral/metabolismo , Genes Virais , Genoma Viral , Fases de Leitura Aberta , Mapeamento por Restrição , Salmonella/virologia , Fagos de Salmonella/crescimento & desenvolvimento , Fagos de Salmonella/patogenicidade , Proteínas Virais/biossíntese , Proteínas Virais/química , Proteínas Virais/genética , Virulência
8.
J Biol Chem ; 283(5): 2724-33, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18045875

RESUMO

Macrophages are a central arm of innate immune defense against intracellular pathogens. They internalize microbes into phagosomes where the invaders are being killed by oxygen and nitrogen reactive species. Despite this battery of antimicrobial molecules, some are able to thrive within the phagosome thus termed intraphagosomal pathogens among which are Salmonella, Leishmania, and Mycobacteria. In mice, a single dominant gene termed Nramp1/Slc11a1 controls innate resistance to such pathogens. This gene is expressed exclusively in myeloid cells. Previously, we have shown that the restricted expression of Nramp1 is regulated by a myeloid cell-specific transcription factor termed IRF-8/ICSBP. It is demonstrated here that the induction of Nramp1 expression in activated macrophages is accompanied by a promoter shift from a repression state elicited by c-Myc to an activation state elicited by the induction of IRF-8 in activated macrophages. This transition from repression to activation is facilitated by a competitive protein-protein interaction with the transcription factor Miz-1. To show that IRF-8 is directly involved in the elimination of intraphagosomal pathogens through the regulation of Nramp1 gene expression, we bred wild type as well as IRF-8 and Nramp1 null mouse strains and examined macrophages derived from bone marrow and peritoneum. Our results clearly show that the absence of IRF-8 and Nramp1 leads to the same phenotype; defective killing of intraphagosomal Salmonella enterica serovar typhimurium and Mycobacterium bovis. Thus, interplay between repression and activation state of the Nramp1 promoter mediated by IRF-8 provides the molecular basis by which macrophages resist intraphagosomal pathogens at early stage after infection.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/imunologia , Imunidade Inata , Fatores Reguladores de Interferon/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Fagossomos/imunologia , Fagossomos/microbiologia , Proteínas Proto-Oncogênicas c-myc/imunologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , DNA/genética , DNA/metabolismo , Primers do DNA/genética , Fatores Reguladores de Interferon/deficiência , Fatores Reguladores de Interferon/genética , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium bovis/imunologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Salmonella typhimurium/imunologia , Transativadores/genética , Transativadores/metabolismo
9.
Dig Dis Sci ; 52(7): 1632-5, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17385031

RESUMO

Crohn's disease (CD) is a chronic multifactorial inflammatory disease. The prevalence of CD in Ashkenazi Jews is higher than in Sephardic Jews. SLC11A1, also known as Nramp1, is a divalent cation antiporter essential for the elimination of intraphagosomal pathogens. SLC11A1 has seven alleles in the promoter region and previous studies have suggested an association between CD and SLC11A1. The aim of this study was to check for a possible association between SLC11A1 promoter alleles and CD in Ashkenazi Jewish patients. DNA samples from healthy Ashkenazi donors and Ashkenazi CD patients were obtained and analyzed for SLC11A1 promoter polymorphism by PCR and DNA sequencing. One hundred thirty-one samples from healthy donors and 131 samples from CD patients were analyzed. Four alleles were identified: approximately 70% of the samples carried allele 3; approximately 30%, allele 2; approximately 1%, allele 1; and <1%, allele 5. There was no difference in allele frequencies between healthy donors and CD patients. No correlation was found between mutations in NOD2/CARD15 and the phenotype of CD. We conclude that the difference in SLC11A1 promoter polymorphism plays no role in CD in Ashkenazi Jews.


Assuntos
Proteínas de Transporte de Cátions/genética , Doença de Crohn/genética , Regiões Promotoras Genéticas , Alelos , Estudos de Casos e Controles , Colite Ulcerativa/genética , Feminino , Humanos , Israel , Masculino , Polimorfismo Genético
10.
J Biol Chem ; 282(8): 5633-40, 2007 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-17189268

RESUMO

Interferon (IFN) regulatory factor-8 (IRF-8), previously known as ICSBP, is a myeloid cell essential transcription factor. Mice with null mutation in IRF-8 are defective in the ability of myeloid progenitor cells to mature toward macrophage lineage. Accordingly, these mice develop chronic myelogenous leukemia (CML). We demonstrate here that IRF-8 is an obligatory regulator of the promyelocytic leukemia (PML) gene in activated macrophages, leading to the expression of the PML-I isoform. This regulation is most effective together with two other transcription factors, IRF-1 and PU.1. PML is a tumor suppressor gene that serves as a scaffold protein for nuclear bodies. IRF-8 is not only essential for the IFN-gamma-induced expression of PML in activated macrophages but also for the formation of nuclear bodies. Reduced IRF-8 transcript levels were reported in CML patients, and a recovery to normal levels was observed in patients in remission following treatment with IFN-alpha. We demonstrate a significant correlation between the levels of IRF-8 and PML in these CML patients. Together, our results indicate that some of the myeloleukemia suppressor activities of IRF-8 are mediated through the regulation of PML. When IRF-8 levels are compromised, the reduced PML expression may lead to genome instability and eventually to the leukemic phenotype.


Assuntos
Regulação Leucêmica da Expressão Gênica , Instabilidade Genômica , Fatores Reguladores de Interferon/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Células Progenitoras Mieloides/metabolismo , Animais , Feminino , Regulação Leucêmica da Expressão Gênica/genética , Instabilidade Genômica/genética , Humanos , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Corpos de Inclusão Intranuclear/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Mutantes , Células Progenitoras Mieloides/patologia , Células NIH 3T3 , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Isoformas de Proteínas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética
11.
Biochem Biophys Res Commun ; 332(1): 271-8, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15896327

RESUMO

Vascular endothelial growth factor (VEGF) is a potent angiogenic factor that has a pivotal role in normal and pathological angiogenesis. VEGF has a long 5' untranslated region harboring an open reading frame (ORF) initiated by a CUG codon that is in-frame with the VEGF coding region. The ORF translation leads to the expression of a long isoform termed L-VEGF that is extended by an additional 180 amino acids. In this communication, we provide evidence that L-VEGF is subjected to proteolytic cleavage leading to the detachment of the 180 aa extension from the VEGF moiety. Using immunofluorescence staining, we show that upon hypoxia this 180 aa extension translocates to the nuclei of expressing cells. Accordingly, immunohistochemical staining of both normal and tumor tissue samples demonstrated restricted nuclear localization of the ORF, which was correlated with cytoplasmic localization of VEGF. This suggests that the 180 aa ORF is involved in VEGF-mediated angiogenic processes.


Assuntos
Hipóxia Celular , Núcleo Celular/metabolismo , Rim/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Transporte Ativo do Núcleo Celular , Linhagem Celular , Núcleo Celular/ultraestrutura , Humanos , Rim/irrigação sanguínea , Rim/citologia , Neoplasias/patologia , Neovascularização Patológica/patologia
12.
Int Immunol ; 15(7): 807-15, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12807819

RESUMO

IFN consensus sequence-binding protein (ICSBP) is a member of the IFN-regulatory factors (IRF) and is thus also called IRF-8. Its expression is restricted to hematopoietic cells and IRF-8\ICSBP(-/-) mice are defective in myeloid cell differentiation. This factor exerts its transcriptional activity through interaction with other transcription factors, which leads to either repression or activation. In this paper, we describe the use of a dominant-negative (DN) mutant of IRF-8\ICSBP designed to serve as a molecular tool to dissociate the role of the various protein-protein interactions. This DN-ICSBP is truncated at the DNA-binding domain and can still associate with other factors, but the heterocomplexes produced are incapable of binding to the DNA. We show that the DN-ICSBP is able to compete for the interaction of IRF-8\ICSBP with either IRF or non-IRF members such as PU.1. Accordingly, this DN construct was able to inhibit the PU.1-dependent expression of the IgLlambda in the plasmacytoma cell line J558L. However, stable expression of this DN-ICSBP led to apoptosis of only hematopoietic cells. The data suggests that DN-ICSBP can form heterocomplexes with an as-yet unidentified survival factor for hematopoietic cells.


Assuntos
Apoptose , Linfócitos B/metabolismo , Proteínas Repressoras/genética , Animais , Linfócitos B/imunologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Vetores Genéticos , Humanos , Cadeias lambda de Imunoglobulina/imunologia , Cadeias lambda de Imunoglobulina/metabolismo , Fatores Reguladores de Interferon , Camundongos , Plasmocitoma , Proteínas Repressoras/metabolismo , Retroviridae/genética , Deleção de Sequência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
J Biol Chem ; 278(45): 44025-32, 2003 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-12904288

RESUMO

Natural resistance-associated macrophage protein 1 (Nramp1) is a proton/divalent cation antiporter exclusively expressed in monocyte/macrophage cells with a unique role in innate resistance to intraphagosomal pathogens. In humans, it is linked to several infectious diseases, including leprosy, pulmonary tuberculosis, visceral leishmaniasis, meningococcal meningitis, and human immunodeficiency virus as well as to autoimmune diseases such as rheumatoid arthritis and Crohn's disease. Here we demonstrate that the restricted expression of Nramp1 is mediated by the macrophage-specific transcription factor IRF-8. This factor exerts its activity via protein-protein interaction, which facilitates its binding to target DNA. Using yeast two-hybrid screen we identified Myc Interacting Zinc finger protein 1 (Miz-1) as new interacting partner. This interaction is restricted to immune cells and takes place on the promoter Nramp1 in association with PU.1, a transcription factor essential for myelopoiesis. Consistent with these data, IRF-8 knockout mice are sensitive to a repertoire of intracellular pathogens. Accordingly, IRF-8-/- mice express low levels of Nramp1 that can not be induced any further. Thus, our results explain in molecular terms the role of IRF-8 in conferring innate resistance to intracellular pathogens and point to its possible involvement in autoimmune diseases.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/fisiologia , Proteínas de Ligação a DNA/farmacologia , Imunidade Inata , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Repressoras/farmacologia , Transativadores/farmacologia , Animais , Doenças Autoimunes , Células COS , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Sinergismo Farmacológico , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Células HL-60 , Humanos , Fatores Reguladores de Interferon , Interferon gama/farmacologia , Fatores de Transcrição Kruppel-Like , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Células NIH 3T3 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Recombinantes de Fusão , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Saccharomyces cerevisiae/genética , Transativadores/genética , Transativadores/fisiologia , Fatores de Transcrição , Transfecção , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA