Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Eur J Neurol ; 27(12): 2662-2667, 2020 12.
Article in English | MEDLINE | ID: mdl-32896048

ABSTRACT

BACKGROUND AND PURPOSE: Mutations in the early growth response 2 gene (EGR2) cause demyelinating, but also axonal, neuropathies differing in severity and age of onset. Except for one family, all reported cases have autosomal dominant inheritance and mutations are localized within the three zinc finger (ZNF) DNA-binding domain. The aim of this study was to provide a clinical and molecular analysis of a novel recessive mutation in EGR2. METHODS: Clinical and electrophysiological assessments of three affected patients, from a consanguineous family, were performed. Genetic analyses of EGR2 were carried out by Sanger sequencing. Functional effects of clinical recessive mutations were assessed using a mammalian two-hybrid assay. RESULTS: A novel missense mutation (c.791C>T; p.P264L) in the homozygous state was detected outside the ZNF domains of the EGR2 gene. Three affected siblings presented with distal demyelinating polyneuropathy with severe sensory loss, progressive thoracolumbar scoliosis and trigeminal neuralgia. Respiratory compromise and cranial nerve dysfunction were also found. Our data indicate that the p.P264L mutation prevents interaction of EGR2 transcription factor with NAB corepressors, suggesting that a disruption of the NAB-EGR2 protein interactions can result in dramatic neuropathy. CONCLUSION: Mutations in, or next to, the R1 domain of EGR2 should be considered with extreme caution for genetic counseling, since these could cause a severe neuropathy with an autosomal recessive manner of transmission.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Early Growth Response Protein 2/genetics , Animals , Homozygote , Humans , Mutation , Transcription Factors/genetics
2.
Prostate ; 73(16): 1721-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23999928

ABSTRACT

BACKGROUND: Increasing age is a significant risk factor for prostate cancer. The prostate is exposed to environmental and endogenous stress that may underlie this remarkable incidence. DNA methylation, genomic imprinting, and histone modifications are examples of epigenetic factors known to undergo change in the aging and cancerous prostate. In this review we examine the data linking epigenetic alterations in the prostate with aging to cancer development. METHODS: An online search of current and past peer reviewed literature on epigenetic changes with cancer and aging was performed. Relevant articles were analyzed. RESULTS: Epigenetic changes are responsible for modifying expression of oncogenes and tumor suppressors. Several of these changes may represent a field defect that predisposes to cancer development. Focal hypermethylation occurs at CpG islands in the promoters of certain genes including GSTP1, RARß2, and RASSF1A with both age and cancer, while global hypomethylation is seen in prostate cancer and known to occur in the colon and other organs. A loss of genomic imprinting is responsible for biallelic expression of the well-known Insulin-like Growth Factor 2 (IGF2) gene. Loss of imprinting (LOI) at IGF2 has been documented in cancer and is also known to occur in benign aging prostate tissue marking the presence of cancer. Histone modifications have the ability to dictate chromatin structure and direct gene expression. CONCLUSIONS: Epigenetic changes with aging represent molecular mechanisms to explain the increased susceptibly of the prostate to develop cancer in older men. These changes may provide an opportunity for diagnostic and chemopreventive strategies given the epigenome can be modified.


Subject(s)
Aging/genetics , Epigenesis, Genetic/genetics , Genetic Predisposition to Disease/genetics , Prostatic Neoplasms/genetics , CpG Islands/genetics , DNA Methylation/genetics , Genomic Imprinting/genetics , Histones/genetics , Humans , Insulin-Like Growth Factor II/genetics , Male , Prostatic Neoplasms/epidemiology , Risk Factors
3.
Vet Immunol Immunopathol ; 119(3-4): 214-21, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17629954

ABSTRACT

Dysregulation of immune responses within joints plays an important role in development of inflammatory arthritis. We determined expression of a panel of immune response and matrix turnover genes in synovial fluid collected from a group of dogs with stifle oligoarthritis and associated degenerative cranial cruciate ligament (CCL) rupture (n=27). We also studied synovial fluid gene expression in dogs affected with other forms of degenerative arthritis (n=9) and in the stifle joint of healthy dogs with intact CCL (n=14). After collection, synovial cells were pelleted and RNA was isolated. Relative expression of cathepsin K, cathepsin S, tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinase-9 (MMP-9), invariant chain (li), toll-like receptor-2 (TLR-2), and TLR-9 was determined using real-time quantitative RT-PCR. Data were normalized to peripheral blood mononuclear cells (PBMC) as an internal control. Relative expression of cathepsin K, MMP-9, TRAP, and li was increased in the stifle synovial fluid of dogs with oligoarthritis, when compared with the stifles of healthy dogs (P<0.05). In contrast, relative expression of all of the genes-of-interest in synovial fluid from joints affected with other forms of arthritis was not significantly different from the stifles of healthy dogs. TRAP expression was also significantly increased in the stifle joints of dogs with oligoarthritis, when compared to joint expression of TRAP in dogs with other forms of degenerative arthritis (P<0.05). In the dogs with stifle oligoarthritis, expression of both matrix turnover and immune response genes was increased in stifle synovial fluid, when compared with the internal PBMC control, whereas in healthy dogs and dogs with other forms of arthritis, only expression of matrix turnover genes was increased in synovial fluid, when compared with the internal PBMC control (P<0.05). Taken together, these findings suggest that antigen-specific immune responses within the stifle joint may be involved in the pathogenesis of persistent synovitis and associated joint degradation in dogs with oligoarthritis and degenerative CCL rupture.


Subject(s)
Anterior Cruciate Ligament Injuries , Arthritis/veterinary , Dog Diseases/genetics , Dog Diseases/immunology , Histocompatibility Antigens/genetics , Rupture/veterinary , Stifle/metabolism , Animals , Anterior Cruciate Ligament/metabolism , Arthritis/genetics , Arthritis/immunology , Dogs , Female , Gene Expression Regulation , Histocompatibility Antigens/metabolism , Male , Reverse Transcriptase Polymerase Chain Reaction , Rupture/genetics , Rupture/immunology , Stifle/immunology
4.
Neuron ; 30(2): 355-68, 2001 May.
Article in English | MEDLINE | ID: mdl-11394999

ABSTRACT

The identification of EGR2 mutations in patients with neuropathies and the phenotype Egr2/Krox20(-/-) have demonstrated that the Egr2 transcription factor is critical for peripheral nerve myelination. However, the mechanism by which these mutations cause disease remains unclear, as most patients present with disease in the heterozygous state, whereas Egr2(+/-) mice are phenotypically normal. To understand the effect of aberrant Egr2 activity on Schwann cell gene expression, we performed microarray expression profiling to identify genes regulated by Egr2 in Schwann cells. These include genes encoding myelin proteins and enzymes required for synthesis of normal myelin lipids. Using these newly identified targets, we have shown that neuropathy-associated EGR2 mutants dominant-negatively inhibit wild-type Egr2-mediated expression of essential myelin genes to levels sufficiently low to result in the abnormal myelination observed in these patients.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Myelin Proteins/genetics , Nerve Tissue Proteins/genetics , Schwann Cells/physiology , Sciatic Nerve/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Early Growth Response Protein 2 , Genes, Reporter , Green Fluorescent Proteins , Humans , Immediate-Early Proteins/genetics , Luminescent Proteins/genetics , Male , Myelin Sheath/genetics , Myelin Sheath/physiology , Nerve Crush , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/cytology , Sciatic Nerve/cytology , Transfection
5.
Biochem Biophys Res Commun ; 283(2): 480-6, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11327726

ABSTRACT

Effective tissue repair results from a rapid, temporally orchestrated series of events. At the site of local tissue injury, the production of many growth factors and cytokines is, in part, stimulated by the early growth response transcription factors such as Egr-1. Egr-1 protein binds to a family of corepressor proteins called NAB which function to block or limit Egr-1 trans-activation of cognate target genes. NAB2 blocks Egr-1 activation of the tissue factor (TF) promoter, Egr-1 stimulated production of PDGF-AB, HGF, TGFbeta(1), and VEGF and the endogenous expression of PDGF-AB and TGFbeta(1). Expression of a wild-type NAB2 but not a dominant negative NAB2 mutant abrogates Egr-1 driven TF promoter activity and tubule formation in an in vitro model of angiogenesis. These findings may have importance in any tissue that is subject to scarring after acute or chronic injury.


Subject(s)
DNA-Binding Proteins/metabolism , Growth Substances/biosynthesis , Immediate-Early Proteins , Neoplasm Proteins , Neovascularization, Physiologic/drug effects , Repressor Proteins/pharmacology , Transcription Factors/metabolism , Cells, Cultured , Early Growth Response Protein 1 , Gene Expression/drug effects , Humans , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Thromboplastin/genetics , Wound Healing/drug effects
6.
Am J Physiol Lung Cell Mol Physiol ; 280(3): L547-54, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11159039

ABSTRACT

Calcitonin gene-related peptide (CGRP) is a potent vasodilator. Our group has reported that exogenous CGRP may prevent or reverse hypoxic pulmonary hypertension in rats. The vasodilatory action of CGRP is mediated primarily by CGRP1 receptors. The calcitonin receptor-like receptor (CRLR) and the orphan receptor RDC-1 have been proposed as CGRP1 receptors, and recent evidence suggests that CRLR can function as either a CGRP1 receptor or an adrenomedullin (ADM) receptor. Receptor activity-modifying proteins (RAMPs) determine the ligand specificity of CRLR: coexpression of CRLR and RAMP1 results in a CGRP1 receptor, whereas coexpression of CRLR and RAMP2 or -3 results in an ADM receptor. We used qualitative, semiquantitative, and real-time quantitative RT-PCR to detect and quantitate the relative expression of these agents in the lungs of rats exposed to normoxia (n = 3) and 1 and 2 wk of chronic hypobaric hypoxia (barometric pressure 380 mmHg, equivalent to an inspired O(2) level of 10%; n = 3/time period). Our results show upregulation of RDC-1, RAMP1, and RAMP3 mRNAs in hypoxic rat lung and no change in CRLR and RAMP2 mRNAs. These findings support a functional role for CGRP and ADM receptors in regulating the adult pulmonary circulation.


Subject(s)
Hypoxia/metabolism , Lung/metabolism , Membrane Proteins/genetics , RNA, Messenger/metabolism , Receptors, Calcitonin Gene-Related Peptide/genetics , Receptors, Cell Surface/genetics , Receptors, Chemokine , Receptors, G-Protein-Coupled , Animals , Base Sequence/genetics , Computer Systems , Intracellular Signaling Peptides and Proteins , Male , Molecular Sequence Data , Protein Isoforms/genetics , Rats , Rats, Sprague-Dawley , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Protein 3 , Receptor Activity-Modifying Proteins , Receptors, Adrenomedullin , Receptors, CXCR , Receptors, Peptide/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Nat Med ; 7(1): 101-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135623

ABSTRACT

The transcription factor early growth response protein 1 (EGR1) is overexpressed in a majority of human prostate cancers and is implicated in the regulation of several genes important for prostate tumor progression. Here we have assessed the effect of Egr1 deficiency on tumor development in two transgenic mouse models of prostate cancer (CR2-T-Ag and TRAMP). Using a combination of high-resolution magnetic resonance imaging and histopathological and survival analyses, we show that tumor progression was significantly impaired in Egr1-/- mice. Tumor initiation and tumor growth rate were not affected by the lack of Egr1; however, Egr1 deficiency significantly delayed the progression from prostatic intra-epithelial neoplasia to invasive carcinoma. These results indicate a unique role for Egr1 in regulating the transition from localized, carcinoma in situ to invasive carcinoma.


Subject(s)
DNA-Binding Proteins/physiology , Immediate-Early Proteins , Neoplasm Proteins , Prostatic Neoplasms/genetics , Transcription Factors/physiology , Animals , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Gene Expression Regulation, Neoplastic , Male , Mice , Mice, Transgenic , Precancerous Conditions/pathology , Repressor Proteins/physiology , Transcription Factors/genetics
8.
J Biol Chem ; 275(49): 38524-31, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-10984481

ABSTRACT

The EGR1 transactivator is overexpressed in prostate cancer, and its expression pattern suggests that EGR1 could potentially regulate a number of steps involved in initiation and progression of prostate cancer, such as mitogenesis, invasiveness, angiogenesis, and metastasis. To identify potential EGR1 target genes in an unbiased manner, we have utilized adenovirus-mediated expression of EGR1 in a prostate cancer cell line to identify specific genes that are induced by EGR1. Using oligonucleotide arrays, a number of EGR1-regulated genes were identified and their regulation was confirmed by quantitative reverse transcription-polymerase chain reaction analysis. One of the largest gene classes identified in this screen includes several neuroendocrine-associated genes (neuron-specific enolase, neurogranin), suggesting that EGR1 overexpression may contribute to the neuroendocrine differentiation that often accompanies prostate cancer progression. This screen also identified several growth factors such as insulin-like growth factor-II, platelet-derived growth factor-A, and transforming growth factor-beta1, which have previously been implicated in enhancing tumor progression. The insulin-like growth factor-II gene lies within the 11p15.5 chromosomal locus, which contains a number of other imprinted genes, and EGR1 expression was found to induce at least two other genes in this locus (IPL, p57(KIP2)). Based on our results, coupling adenoviral overexpression with microarray and quantitative reverse transcription-polymerase chain reaction analyses could be a versatile strategy for identifying target genes of transactivators.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/genetics , Prostatic Neoplasms/genetics , Transcription Factors/metabolism , Alleles , Amino Acid Substitution , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Enzymes/genetics , Humans , Immediate-Early Proteins/metabolism , Male , Oligonucleotide Array Sequence Analysis , Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
9.
J Biol Chem ; 275(13): 9749-57, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734128

ABSTRACT

The EGR1/NGFI-A transcription factor directly activates the luteinizing hormone beta (LHbeta) subunit promoter, and female mice lacking EGR1 are infertile due to LHbeta deficiency. The NGFI-A-binding proteins NAB1 and NAB2 are corepressors of EGR1/NGFI-A and of the related proteins EGR2/Krox20 and EGR3. Here we report that at certain promoters, including LHbeta, NAB proteins display a novel ability to stimulate EGR-directed transcription. NAB coactivation requires the conserved NCD2 protein domain, previously implicated in NAB corepression, is strictly dependent upon EGR binding to the LHbeta proximal promoter and is independent of EGR activation domains. Furthermore, we report that NAB-activated promoters such as LHbeta contain EGR consensus sites that are fewer in number and lower in binding affinity than those found at NAB-repressed promoters such as basic fibroblast growth factor. Analysis of mutant and synthetic promoters confirms that both the strength and multiplicity of EGR-binding sites influence the transcriptional outcome of NAB recruitment. These results suggest a novel means by which EGR target genes could be differentially regulated in cells where EGR and NAB proteins are coexpressed.


Subject(s)
DNA-Binding Proteins/physiology , Immediate-Early Proteins , Luteinizing Hormone/genetics , Neoplasm Proteins , Repressor Proteins/physiology , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , Binding Sites , DNA Primers , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Female , Mice , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism
10.
J Biol Chem ; 275(12): 9035-42, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722753

ABSTRACT

Chromatin reorganization of the PHO5 and murine mammary tumor virus (MMTV) promoters is triggered by binding of either Pho4 or the glucocorticoid receptor (GR), respectively. In order to compare the ability of Pho4 and GR to remodel chromatin and activate transcription, hybrid promoter constructs were created by insertion of the MMTV B nucleosome sequence into the PHO5 promoter and then transformed into a yeast strain expressing GR. Activation of either Pho4 (by phosphate depletion) or GR (by hormone addition) resulted in only slight induction of hybrid promoter activity. However, simultaneous activation of both Pho4 and GR resulted in synergistic activation to levels exceeding that of the wild type PHO5 promoter. Under these conditions, Pho4 completely disrupted the nucleosome containing its binding site. In contrast, GR had little effect on the stability of the MMTV B nucleosome. A minimal transactivation domain of the GR fused to the Pho4 DNA-binding domain is capable of efficiently disrupting the nucleosome with a Pho4-binding site, whereas the complementary hybrid protein (Pho4 activation domain, GR DNA-binding domain) does not labilize the B nucleosome. Therefore, we conclude that significant activation by Pho4 requires nucleosome disruption, whereas equivalent transcriptional activation by GR is not accompanied by overt perturbation of nucleosome structure. Our results show that the DNA-binding domains of the two factors play critical roles in determining how chromatin structure is modified during promoter activation.


Subject(s)
DNA-Binding Proteins , Fungal Proteins/metabolism , Nucleosomes/metabolism , Receptors, Glucocorticoid/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , Chromatin/metabolism , Chromatin/ultrastructure , Mammary Tumor Virus, Mouse/genetics , Nucleosomes/ultrastructure , Promoter Regions, Genetic , Protein Binding , Transcriptional Activation
11.
Hum Mol Genet ; 8(7): 1245-51, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10369870

ABSTRACT

The early growth response 2 gene ( EGR2 ) is a Cys2His2zinc finger transcription factor which is thought to play a role in the regulation of peripheral nervous system myelination. This idea is based partly on the phenotype of homozygous Krox20 ( Egr2 ) knockout mice, which display hypomyelination of the PNS and a block of Schwann cells at an early stage of differentiation. Mutations in the human EGR2 gene have recently been associated with the inherited peripheral neuropathies Charcot-Marie-Tooth type 1, Dejerine-Sottas syndrome and congenital hypomyelinating neuropathy. Three of the four EGR2 mutations are dominant and occur within the zinc finger DNA-binding domain. The fourth mutation is recessive and affects the inhibitory domain (R1) that binds the NAB transcriptional co-repressors. A combination of DNA-binding assays and transcriptional analysis was used to determine the functional consequences of these mutations. The zinc finger mutations affect DNA binding and the amount of residual binding directly correlates with disease severity. The R1 domain mutation prevents interaction of EGR2 with the NAB co-repressors and thereby increases transcriptional activity. These data provide insight into the possible disease mechanisms underlying EGR2 mutations and the reason for varying severity and differences in inheritance patterns.


Subject(s)
DNA-Binding Proteins/genetics , Demyelinating Diseases/genetics , Mutation , Transcription Factors/genetics , Animals , Cells, Cultured , Charcot-Marie-Tooth Disease/genetics , Chlorocebus aethiops , Early Growth Response Protein 2 , Expressed Sequence Tags , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Transcriptional Activation , Zinc Fingers/genetics
12.
J Biol Chem ; 274(20): 13870-6, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318795

ABSTRACT

We have previously shown that early growth response (Egr) 1-deficient mice exhibit female infertility, reflecting a luteinizing hormone (LH) beta deficiency. Egr-1 activates the LHbeta gene in vitro through synergy with steroidogenic factor-1 (SF-1), a protein required for gonadotrope function. To test if this synergy is essential for gonadotropin-releasing hormone (GnRH) stimulation of LHbeta, we examined the activity of the LHbeta promoter in the gonadotrope cell line LbetaT2. GnRH markedly stimulated the LHbeta promoter (15-fold). Mutation of either Egr-1 or SF-1 elements within the LHbeta promoter attenuated this stimulation, whereas mutation of both promoter elements abrogated GnRH induction of the LHbeta promoter. Furthermore, GnRH stimulated Egr-1 but not SF-1 expression in LbetaT2 cells. Importantly, overexpression of Egr-1 alone was sufficient to enhance LHbeta expression. Although other Egr proteins are expressed in LbetaT2 cells and are capable of interacting with SF-1, GnRH stimulation of Egr-1 was the most robust. We also found that the nuclear receptor DAX-1, a repressor of SF-1 activity, reduced Egr-1-SF-1 synergy and diminished GnRH stimulation of the LHbeta promoter. We conclude that the synergy between Egr-1 and SF-1 is essential for GnRH stimulation of the LHbeta gene and plays a central role in the dynamic regulation of LHbeta expression.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Gonadotropin-Releasing Hormone/physiology , Immediate-Early Proteins/physiology , Luteinizing Hormone/genetics , Repressor Proteins , Transcription Factors/physiology , Zinc Fingers , Animals , Cell Line , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/metabolism , Drug Synergism , Early Growth Response Protein 1 , Female , Fushi Tarazu Transcription Factors , Homeodomain Proteins , Mice , Promoter Regions, Genetic , Pulsatile Flow , Receptors, Cytoplasmic and Nuclear , Receptors, Retinoic Acid/metabolism , Steroidogenic Factor 1 , Transcription Factors/metabolism , Transfection
13.
EMBO J ; 17(20): 6010-9, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9774344

ABSTRACT

The NGFI-A binding corepressors NAB1 and NAB2 interact with a conserved domain (R1 domain) within the Egr1/NGFI-A and Egr2/Krox20 transactivators, and repress the transcription of Egr target promoters. Using a novel adaptation of the yeast two-hybrid screen, we have identified several point mutations in NAB corepressors that interfere with their ability to bind to the Egr1 R1 domain. Surprisingly, NAB proteins bearing some of these mutations increased Egr1 activity dramatically. The mechanism underlying the unexpected behavior of these mutants was elucidated by the discovery that NAB conserved domain 1 (NCD1) not only binds to Egr proteins but also mediates multimerization of NAB molecules. The activating mutants exert a dominant negative effect on NAB repression by multimerizing with native NAB proteins and preventing binding of endogenous NAB proteins with Egr transactivators. To examine NAB repression of a native Egr target gene, we show that NAB2 represses Egr2/Krox20-mediated activation of the bFGF/FGF-2 promoter, and that repression is reversed by coexpression of dominant negative NAB2. Because of their specific ability to alleviate NAB repression of Egr target genes, the dominant negative NAB mutants will be useful in elucidating the mechanism and function of NAB corepressors.


Subject(s)
Mutagenesis, Site-Directed , Neoplasm Proteins , Repressor Proteins/genetics , Trans-Activators/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Cells, Cultured , Chlorocebus aethiops , Conserved Sequence , Genes, Helminth , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Peptide Fragments/genetics , Peptide Fragments/physiology , Precipitin Tests , Protein Binding/genetics , Rats , Repressor Proteins/physiology
14.
Mol Cell Biol ; 18(10): 5818-27, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742099

ABSTRACT

Perhaps the best characterized example of an activator-induced chromatin transition is found in the activation of the Saccharomyces cerevisiae acid phosphatase gene PHO5 by the basic helix-loop-helix (bHLH) transcription factor Pho4. Transcription activation of the PHO5 promoter by Pho4 is accompanied by the remodeling of four positioned nucleosomes which is dependent on the Pho4 activation domain but independent of transcription initiation. Whether the requirements for transcription activation through the TATA sequence are different from those necessary for the chromatin transition remains a major outstanding question. In an attempt to understand better the ability of Pho4 to activate transcription and to remodel chromatin, we have initiated a detailed characterization of the Pho4 activation domain. Using both deletion and point mutational analysis, we have defined residues between positions 75 and 99 as being both essential and sufficient to mediate transcription activation. Significantly, there is a marked concordance between the ability of mutations in the Pho4 activation domain to induce chromatin opening and transcription activation. Interestingly, the requirements for transcription activation within the Pho4 activation domain differ significantly if fused to a heterologous bHLH-leucine zipper DNA-binding domain. The implications for transcription activation by Pho4 are discussed.


Subject(s)
Chromatin , DNA-Binding Proteins/physiology , Fungal Proteins/physiology , Helix-Loop-Helix Motifs , Saccharomyces cerevisiae Proteins , Transcription Factors/physiology , Transcriptional Activation , Amino Acid Sequence , Amino Acids , Binding Sites , Circular Dichroism , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Molecular Sequence Data , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Cell Biol ; 142(4): 1075-82, 1998 Aug 24.
Article in English | MEDLINE | ID: mdl-9722618

ABSTRACT

The PC12 pheochromocytoma cell line responds to NGF by undergoing growth arrest and proceeding to differentiate toward a neuronal phenotype. Among the early genetic events triggered by NGF in PC12 cells are the rapid activation of the zinc finger transcription factor Egr1/NGFI-A, and a slightly delayed induction of NAB2, a corepressor that inhibits Egr1 transcriptional activity. We found that stably transfected PC12 cells expressing high levels of NAB2 do not differentiate, but rather continue to proliferate in response to NGF. Inhibition of PC12 differentiation by NAB2 overexpression was confirmed using two additional experimental approaches, transient transfection, and adenoviral infection. Early events in the NGF signaling cascade, such as activation of MAP kinase and induction of immediate-early genes, were unaltered in the NAB2-overexpressing PC12 cell lines. However, induction of delayed NGF response genes such as TGF-beta1 and MMP-3 was inhibited. Furthermore, NAB2 overexpression led to downregulation of p21(WAF1), a molecule previously shown to play a pivotal role in the ability of PC12 cells to undergo growth arrest and commit to differentiation in response to NGF. Cotransfection with p21(WAF1) restored the ability of NAB2-overexpressing PC12 cells to differentiate in response to NGF.


Subject(s)
Cell Differentiation/physiology , Immediate-Early Proteins , Neoplasm Proteins , Nerve Growth Factors/pharmacology , Repressor Proteins/pharmacology , Repressor Proteins/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Division/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/physiology , Early Growth Response Protein 1 , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , Genes, Reporter/genetics , Matrix Metalloproteinase 3/metabolism , PC12 Cells , Rats , Transcription Factors/physiology , Transfection/genetics , Transforming Growth Factor beta/metabolism
16.
Mol Cell Biol ; 18(1): 512-24, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418898

ABSTRACT

Nab proteins constitute an evolutionarily conserved family of corepressors that specifically interact with and repress transcription mediated by three members of the NGFI-A (Egr-1, Krox24, zif/268) family of immediate-early gene transcription factors, which includes NGFI-C, Krox20, and Egr3. We explored the mechanism of Nab1 repression and identified structural domains required for Nab1 function. Nab1 does not act by blocking DNA binding or nuclear localization of NGFI-A. In fact, Nab1 repression is not unique to NGFI-A because multiple types of non-NGFI-A activation domains were repressed, as was a heterologous transcription factor carrying the NGFI-A R1 domain, which is required for Nab1 interaction. Additionally, Nab1 tethered directly to DNA repressed constitutively active promoters. Tethered repression was not dependent on the identity of the basal promoter elements, the presence of a distal enhancer, or the distance separating the binding sites from the promoter. These results suggest that Nab1 repression is not specific to particular activators and that Nab1 is an active repressor that works by a direct mechanism. We identified a bipartite-like nuclear localization sequence and localized the repression function to the Nab conserved domain 2 (NCD2), a region found in the carboxy-terminal half of all Nab proteins. Three small regions of homology between Nab1 and previously characterized corepressors, Dr1 and E1b 55-kDa protein, were identified within NCD2. Replacement mutagenesis of residues conserved between these proteins interfered with Nab1 repression, although Nab1 does not function by the same mechanism as Dr1. The human NAB1 genomic locus was mapped to chromosome 2q32.3-33.


Subject(s)
Chromosomes, Human, Pair 2 , DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Immediate-Early Proteins , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins , Transcription Factors/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , COS Cells , Chromosome Mapping , Early Growth Response Protein 1 , Humans , Molecular Sequence Data , RNA-Binding Proteins/genetics , Sequence Analysis , Transcriptional Activation , Zinc Fingers
17.
Genomics ; 41(1): 33-9, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9126479

ABSTRACT

The two Nab genes, coding for transcriptional corepressors of NGFI-A (Egr-1, Krox24, zif268) and Krox20, have been localized to two regions of the genome, each of which contains at least two members of the Stat gene family. The association of the two Nab genes with the Stat clusters on mouse chromosomes 1 and 10 (human chromosomes 2 and 12) suggest that a Nab gene was involved in at least one of the duplication events that resulted in dispersion of the primordial Stat gene pair to three different mouse chromosomes. Sequencing of the Nab2 genomic locus revealed that it is situated very close to the Stat6 gene. The transcripts of the two genes converge, such that the 3' ends of the Stat6 and Nab2 mRNAs overlap by 58 bp. Both transcripts terminate within a 78-bp region that is absolutely conserved between mouse and human. Analysis of Nab2 cDNA revealed that there is an alternatively spliced form of the Nab2 transcript (lacking exon 3) that produces a protein that lacks the ability to repress transcription by NGFI-A and Krox20.


Subject(s)
Neoplasm Proteins , Repressor Proteins/genetics , Trans-Activators/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Conserved Sequence , DNA Primers/genetics , DNA, Complementary/genetics , Humans , Mice , Molecular Sequence Data , Multigene Family , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , STAT6 Transcription Factor , Species Specificity , Trans-Activators/metabolism , Transcription, Genetic
18.
Trends Biochem Sci ; 22(3): 93-7, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9066259

ABSTRACT

Activation of the Saccharomyces cerevisiae PHO5 gene is accompanied by the disruption of four positioned nucleosomes at the promoter. The chromatin transition requires a DNA-binding protein, Pho4, and its transactivation domain. The mechanism of nucleosome disruption and the contribution of the nucleosomes to PHO5 regulation are reviewed.


Subject(s)
Gene Expression Regulation, Fungal , Membrane Transport Proteins/genetics , Nucleosomes/metabolism , Phosphate Transport Proteins , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Chromatin/chemistry , Chromatin/metabolism , Chromatin/ultrastructure , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription, Genetic
19.
Nucleic Acids Res ; 24(22): 4479-86, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8948638

ABSTRACT

Two transcription factors, the bHLH protein Pho4 and the homeodomain protein Pho2, are required for transcriptional activation of the PHO5 promoter in Saccharomyces cerevisiae. There are two essential Pho4 binding sites, corresponding to the regulatory elements UASp1 and UASp2 at the PHO5 promoter, but only a single, dispensable Pho2 binding site had previously been identified. We have reinvestigated binding of Pho2 to the PHO5 promoter using purified recombinant protein and have found multiple Pho2 binding sites of different affinities along the promoter. One of the high affinity Pho2 sites largely overlaps the Pho4 binding site at UASp1. Cooperative DNA binding of the two proteins to their overlapping sites, resulting in a high-affinity ternary complex, was demonstrated. Pho2 and Pho4 also bind DNA cooperatively at UASp2 where two Pho2 sites flank the Pho4 site. Finally, Pho2 facilitates binding of Pho4 to a third, cryptic Pho4 binding site which binds Pho4 with lower affinity than UASp1 or UASp2. These results suggest that cooperative DNA binding with Pho4 is integral to the mechanism by which Pho2 regulates transcription of the PHO5 gene.


Subject(s)
DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Helix-Loop-Helix Motifs , Homeodomain Proteins , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites , Molecular Sequence Data
20.
Mol Cell Biol ; 16(7): 3545-53, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668170

ABSTRACT

Previous work had identified a corepressor, NAB1, which represses transcriptional activation mediated by NGFI-A (also known as Egr-1, zif268, and Krox24) and Krox20. These zinc finger transcription factors are encoded by immediate-early genes and have been implicated in a wide variety of proliferative and differentiative processes. We have isolated and characterized another corepressor, NAB2, which is highly related to NAB1 within two discrete domains. The first conserved domain of NAB2 mediates an interaction with the R1 domain of NGFI-A. NAB2 represses the activity of both NGFI-A and Krox20, and its expression is regulated by some of the same stimuli that induce NGFI-A expression, including serum stimulation of fibroblasts and nerve growth factor stimulation of PC12 cells. The human NAB2 gene has been localized to chromosome 12ql3.3-14.1, a region that is rearranged in several solid tumors, lipomas, uterine leiomyomata, and liposarcomas. Sequencing of the Caenorhabditis elegans genome has identified a gene that bears high homology to both NAB1 and NAB2, suggesting that NAB molecules fulfill an evolutionarily conserved role.


Subject(s)
Biological Evolution , Chromosomes, Human, Pair 12 , DNA-Binding Proteins/metabolism , Immediate-Early Proteins , Neoplasm Proteins , Neoplasms/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Caenorhabditis elegans/genetics , Cell Differentiation , Cell Division , Chromosome Mapping , Consensus Sequence , Conserved Sequence , DNA Primers , Early Growth Response Protein 1 , Early Growth Response Protein 2 , Female , Gene Rearrangement , Genome , Humans , In Situ Hybridization, Fluorescence , Leiomyoma/genetics , Lipoma/genetics , Liposarcoma/genetics , Mice , Molecular Sequence Data , Organ Specificity , Placenta/metabolism , Polymerase Chain Reaction , Pregnancy , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Transcriptional Activation , Uterine Neoplasms/genetics , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...