Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cells ; 11(8)2022 04 11.
Article in English | MEDLINE | ID: mdl-35455969

ABSTRACT

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.


Subject(s)
Neovascularization, Pathologic , Vascular Endothelial Growth Factor A , Apoptosis , Cell Hypoxia , Humans , Hypoxia , Neovascularization, Pathologic/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
J Mol Biol ; 432(16): 4544-4560, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32534063

ABSTRACT

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.


Subject(s)
Chromatin/metabolism , Interferon Regulatory Factors/genetics , MafK Transcription Factor/genetics , MafK Transcription Factor/metabolism , Animals , Binding Sites , CRISPR-Cas Systems , Chromatin Assembly and Disassembly , Gene Expression Regulation , HEK293 Cells , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/metabolism , Mice , NIH 3T3 Cells , Organ Specificity , RAW 264.7 Cells , RNA, Small Interfering/pharmacology
3.
J Mol Biol ; 431(2): 210-222, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30502383

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Interferon Regulatory Factors/genetics , Introns/genetics , Mouse Embryonic Stem Cells/metabolism , Animals , Cell Line , Cell Lineage/genetics , Dendritic Cells/metabolism , Gene Expression Regulation/genetics , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Mice , Monocytes/metabolism , Myocytes, Cardiac/metabolism , NIH 3T3 Cells , RAW 264.7 Cells
4.
PLoS One ; 11(6): e0156812, 2016.
Article in English | MEDLINE | ID: mdl-27257682

ABSTRACT

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.


Subject(s)
Chromatin/metabolism , Interferon Regulatory Factors/metabolism , Introns/genetics , Macrophages/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , NIH 3T3 Cells , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction
5.
Int Immunol ; 23(4): 287-96, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21427174

ABSTRACT

IFN regulatory factor-8 (IRF-8, previously known as ICSBP) is a key transcription factor driving the differentiation of granulocyte\monocyte progenitor (GMP) cells toward monocyte\macrophage lineage. The promyelocytic leukemia (PML) gene is an immediate target gene regulated by IRF-8 in response to IFN-γ activation. PML is a multifunctional protein that has many isoforms serving as the scaffold components for nuclear bodies (NBs) engaged in numerous proteins interactions. The role of PML in the retinoic acid pathway that drives GMPs to granulopoiesis is documented in the literature. Here, we show that PML is also involved in monopoiesis by mediating some of the IRF-8 activities during the differentiation of murine-derived bone marrow macrophages (BMMs). PML silencing resulted in altered expression level of key transcription factors essential for monopoiesis that was accompanied by silencing of typical myeloid-specific genes. Interestingly, this altered expression resembled that of the GMPs and that of BMMs derived from IRF-8(-/-) mice altogether supporting the role of PML in monopoiesis. Further, PML silencing led to reduced colony-forming capacity of bone marrow cells highlighting the dual function of PML in myelopoiesis. Last, PML overexpression only partially rescued the phenotype of IRF-8(-/-) BMMs. Together, our data show that PML is an important factor for monopoiesis and not solely for granulopoiesis. This suggests that PML-NBs respond to an incoming signal that affects the fate of GMP driving cell differentiation to granulocytes or monocytes.


Subject(s)
Macrophages/metabolism , Myeloid Progenitor Cells/cytology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Promyelocytic Leukemia Protein , RNA, Small Interfering/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
6.
J Biol Chem ; 283(5): 2724-33, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18045875

ABSTRACT

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.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Immunity, Innate , Interferon Regulatory Factors/immunology , Macrophages/immunology , Macrophages/microbiology , Phagosomes/immunology , Phagosomes/microbiology , Proto-Oncogene Proteins c-myc/immunology , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , DNA Primers/genetics , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/immunology , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Salmonella typhimurium/immunology , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Dig Dis Sci ; 52(7): 1632-5, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17385031

ABSTRACT

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.


Subject(s)
Cation Transport Proteins/genetics , Crohn Disease/genetics , Promoter Regions, Genetic , Alleles , Case-Control Studies , Colitis, Ulcerative/genetics , Female , Humans , Israel , Male , Polymorphism, Genetic
8.
Mol Immunol ; 44(4): 338-46, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16597464

ABSTRACT

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.


Subject(s)
Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factors/genetics , Macrophage Activation/genetics , Animals , Cell Line , Gene Expression Profiling , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/metabolism , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/physiology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Transcriptional Activation/drug effects
9.
J Biol Chem ; 282(8): 5633-40, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17189268

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Leukemic , Genomic Instability , Interferon Regulatory Factors/metabolism , Intranuclear Inclusion Bodies/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloid Progenitor Cells/metabolism , Animals , Female , Gene Expression Regulation, Leukemic/genetics , Genomic Instability/genetics , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/genetics , Intranuclear Inclusion Bodies/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Mutant Strains , Myeloid Progenitor Cells/pathology , NIH 3T3 Cells , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Isoforms , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
10.
J Interferon Cytokine Res ; 25(12): 770-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16375605

ABSTRACT

Both type I interferon (IFN-alpha/beta) and type II IFN (IFN-gamma) exert many functions that are restricted to immune cells. Thus, they play critical roles in innate and adaptive immunity. IFN regulatory factor-4 (IRF-4) and IRF-8 (formerly PU.1 interaction partner [Pip] and IFN consensus sequence binding domain [ICSBP], respectively) are immune cell-specific members of the IRF family that regulate the development of myeloid, lymphoid, and dendritic cells. They form a heterodimeric complex with another immune cell-specific transcription factor PU.1-Spi-1 and regulate transcription of genes in the immune system. This review describes the role of the IRF-8-PU.1 complex in modulating IFN signaling in an immune cell-specific manner. Our studies revealed that some but not all IFN-gamma-inducible genes carry an IFN-gamma activation site (GAS) element that contains a binding site for the IRF- 8-PU.1 complex. The IRF-8-PU.1 complex can take part in GAS-mediated transcription and amplify expression of IFN-gamma-responsive genes initiated by Stat1 in macrophages. Similarly, some but not all IFN-alpha/beta-responsive genes are shown to carry an IFN-stimulated response element (ISRE) that contains an IRF-8-PU.1 binding site. The participation of IRF-8-PU.1 in ISRE-mediated transcription results in the augmentation of IFN-stimulated gene factor 3 (ISGF3)-induced transcription in macrophages. Thus, GAS and ISRE elements, classically defined as universal IFN-alpha/beta and IFN-gamma response sequences, are not the same, and some harbor an embedded motif for IRF- 8-PU.1 binding that functions only in immune cells. Accordingly, the IRF-8-PU.1complex provides secondary IFN signaling pathways unique to the immune system. Collectively, the contribution of IRF-8 and PU.1 to IFN-regulated gene expression may in part account for immune cell-specific functions of IFNs.


Subject(s)
Interferon Regulatory Factors/immunology , Interferons/immunology , Proto-Oncogene Proteins/immunology , Signal Transduction/immunology , Trans-Activators/immunology , Animals , Base Sequence , DNA/genetics , DNA/metabolism , Humans
11.
Biochem Biophys Res Commun ; 332(1): 271-8, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15896327

ABSTRACT

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.


Subject(s)
Cell Hypoxia , Cell Nucleus/metabolism , Kidney/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/metabolism , Active Transport, Cell Nucleus , Cell Line , Cell Nucleus/ultrastructure , Humans , Kidney/blood supply , Kidney/cytology , Neoplasms/pathology , Neovascularization, Pathologic/pathology
12.
Biochem Biophys Res Commun ; 329(4): 1288-92, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15766566

ABSTRACT

The transcription factor PU.1 plays a central role in development and differentiation of hematopoietic cells. Evidence from PU.1 knockout mice indicates a pivotal role for PU.1 in myeloid lineage and B-lymphocyte development. In addition, PU.1 is a key player in the development of Friend erythroleukemia disease, which is characterized by proliferation and differentiation arrest of proerythrocytes. To study the role of PU.1 in erythroleukemia, we have used murine erythroleukemia cells, isolated from Friend virus-infected mice. Expression of PU.1 small interfering RNA in these cells led to significant inhibition of PU.1 levels. This was accompanied by inhibition of proliferation and restoration in the ability of the proerythroblastic cells to produce hemoglobin, i.e., reversion of the leukemic phenotype. The data suggest that overexpression of PU.1 gene is the immediate cause for maintaining the leukemic phenotype of the disease by retaining the self-renewal capacity of transformed erythroblastic cells and by blocking the terminal differentiation program towards erythrocytes.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Neoplastic , Gene Silencing , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Cell Cycle , Cell Line , Hemoglobins/biosynthesis , Hemoglobins/metabolism , Leukemia, Erythroblastic, Acute/genetics , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic/genetics
13.
J Biol Chem ; 278(45): 44025-32, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-12904288

ABSTRACT

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.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/physiology , DNA-Binding Proteins/pharmacology , Immunity, Innate , Proto-Oncogene Proteins/pharmacology , Repressor Proteins/pharmacology , Trans-Activators/pharmacology , Animals , Autoimmune Diseases , COS Cells , Cell Line , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Drug Synergism , Gene Expression/drug effects , Genetic Vectors , HL-60 Cells , Humans , Interferon Regulatory Factors , Interferon-gamma/pharmacology , Kruppel-Like Transcription Factors , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Mice, Knockout , NIH 3T3 Cells , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Recombinant Fusion Proteins , Repressor Proteins/genetics , Repressor Proteins/physiology , Saccharomyces cerevisiae/genetics , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors , Transfection , Two-Hybrid System Techniques
14.
J Biol Chem ; 278(38): 36017-26, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12840021

ABSTRACT

Murine Nramp1 encodes a divalent cation transporter that is expressed in late endosomes/lysosomes of macrophages, and the transported cations facilitate intracellular pathogen growth control. The Nramp1 promoter is TATA box-deficient, has two initiator elements, and is repressed by c-Myc, in accordance with the notion that genes that deplete the iron content of the cell cytosol antagonize cell growth. Repression via c-Myc occurs at the initiator elements, whereas a c-Myc-interacting protein (Miz-1) stimulates transcription. Here we demonstrate that a non-canonical E box (CAACTG) inhibits basal promoter activity and activation by Miz-1. A consensus Sp1-binding site or GC box is also necessary for Miz-1-dependent transactivation, but not repression. Repression occurs by c-Myc competing with p300/CBP for binding Miz-1. Our results show that an Sp1 site mutant inhibits coactivation by p300 and that the murine Nramp1 promoter is preferentially expressed within macrophages (relative to a beta-actin control) compared with non-macrophage cells. The effect of the Sp1 site mutation on promoter function shows cell-type specificity: stimulation in COS-1 and inhibition in RAW264.7 cells. Miz-1-directed RNA interference confirms a stimulatory role for Miz-1 in Nramp1 promoter function. c-Myc, Miz-1, and Sp1 were identified as binding to the Nramp1 core promoter in control cells and following acute stimulation with interferon-gamma and lipopolysaccharide. These results provide a description of sites that modulate the activity of the initiator-binding protein Miz-1 and indicate a stimulatory role for GC box-binding factors in macrophages and a inhibitory role for E box elements in proliferating cells.


Subject(s)
Cation Transport Proteins/genetics , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Animals , Binding Sites , COS Cells , Cations , Cell Division , Chromatin/metabolism , Dose-Response Relationship, Drug , Gene Deletion , Genes, Reporter , Interferon-gamma/metabolism , Iron/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Models, Genetic , Mutation , Plasmids/metabolism , Precipitin Tests , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Transfection , Zinc Fingers
15.
Int Immunol ; 15(7): 807-15, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12807819

ABSTRACT

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.


Subject(s)
Apoptosis , B-Lymphocytes/metabolism , Repressor Proteins/genetics , Animals , B-Lymphocytes/immunology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Genetic Vectors , Humans , Immunoglobulin lambda-Chains/immunology , Immunoglobulin lambda-Chains/metabolism , Interferon Regulatory Factors , Mice , Plasmacytoma , Repressor Proteins/metabolism , Retroviridae/genetics , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism
16.
J Immunol ; 168(12): 6224-31, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12055236

ABSTRACT

Type I IFNs cause the induction of a subset of genes termed IFN-stimulated genes (ISGs), which harbor a specific DNA element, IFN-stimulated response element (ISRE). This ISRE confers the responsiveness to the IFN signal through the binding of a family of transcription factors designated IFN regulatory factors (IRFs). Some IRFs can bind to the DNA alone, such as IRF-1, which elicits transcriptional activation, or IRF-2, which leads to transcriptional repression. In addition, these factors associate with IRF-8/IFN consensus sequence binding protein (ICSBP), an immune cell-restricted IRF, and the assembled heterocomplexes lead to synergistic repression of ISRE elements. ISG15 is a prototype ISG that contains a well-characterized ISRE. Here we show that PU.1, an ETS member essential for myeloid/lymphoid cell differentiation, forms heterocomplexes with the immune-restricted IRFs, IRF-8\/ICSBP and IRF-4, which lead to transcriptional activation of ISG15. These data allowed the characterization of a subset of ISREs designated ETS/IRF response element (EIRE), which are differentially regulated in immune cells. EIREs are unique in their ability to recruit different factors to an assembled enhanceosomes. In nonimmune cells the factors will mainly include IRF members, while cell type-restricted factors, such as PU.1, IRF-8\/ICSBP, and IRF-4, will be recruited in immune cells. IRF heterocomplex formation leads to transcriptional repression, and conversely, PU.1/IRFs heterocomplex formation leads to transcriptional activation. The fact that IRF-8\/ICSBP is an IFN-gamma-induced factor explains why some of the EIREs are also induced by type II IFN. Our results lay the molecular basis for the unique regulation of ISGs, harboring EIRE, in immune cells.


Subject(s)
Cytokines/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/immunology , Interferon Type I/pharmacology , Interferons/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Response Elements/immunology , Trans-Activators/metabolism , Transcription Factors/metabolism , Ubiquitins/analogs & derivatives , 3T3 Cells , Animals , Consensus Sequence/immunology , Cytokines/antagonists & inhibitors , Cytokines/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Drug Synergism , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/metabolism , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factors , K562 Cells/chemistry , K562 Cells/metabolism , Mice , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/physiology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/physiology , Repressor Proteins/biosynthesis , Repressor Proteins/physiology , Trans-Activators/biosynthesis , Trans-Activators/physiology , Transcription Factors/biosynthesis , Transcription Factors/physiology , Transcriptional Activation/immunology , U937 Cells/chemistry , U937 Cells/metabolism
17.
J Interferon Cytokine Res ; 22(1): 153-60, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11846986

ABSTRACT

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.


Subject(s)
Repressor Proteins/metabolism , Transcriptional Activation , Animals , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Interferon Regulatory Factors , Macromolecular Substances , Macrophages/metabolism , Models, Molecular , Molecular Sequence Data , Promoter Regions, Genetic , Protein Structure, Tertiary , Repressor Proteins/chemistry , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...