Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cancer Res ; 61(24): 8803-10, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751402

ABSTRACT

Id helix-loop-helix (HLH) proteins function as regulators of cell growth and differentiation and when overexpressed can induce malignant transformation. In a series of 34 cases of primary human colorectal adenocarcinoma, immunoreactivity for Id1, Id2, and Id3 was found to be significantly elevated in tumor compared with normal mucosa (P = 0.001 for Id1 and Id2; P = 0.002 for Id3). No elevation of Id expression was observed in 17 cases of adenoma. Expression of Id1 and to a lesser extent of Id2 was correlated with mitotic index (P = 0.005 for Id1; P = 0.042 for Id2) in human adenocarcinomas, and expression of all three Id proteins was correlated with p53 immunoreactivity (a marker of mutational 'inactivation' of p53 function; P = 0.002 for Id1; P = 0.006 for Id2; P = 0.016 for Id3). In normal intestinal mucosa of p53-null mice and in spontaneous tumors arising in Min+/- mice, expression of all three Id proteins was also found to be up-regulated. Antisense oligonucleotide blockade of Id protein expression inhibited the proliferation of human adenocarcinoma cells. Enforced, ectopic expression of the E47 basic HLH (bHLH) protein in human adenocarcinoma cell lines efficiently sequestered endogenous Id proteins as Id-bHLH heterodimers, as shown by coimmunoprecipitation and subcellular colocalization studies. This led to growth arrest of the cells. Enforced overexpression of a mutant E47 protein, deficient in transactivation and DNA binding function, also partially inhibited cell growth. Taken together, these data imply that deregulated expression of Id proteins in colorectal adenocarcinoma arises at least in part as a consequence of loss of p53 function and contributes to the uncontrolled proliferation of tumor cells in colorectal cancer.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/biosynthesis , Helix-Loop-Helix Motifs , Repressor Proteins , Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Adenocarcinoma/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Division/drug effects , Cell Division/physiology , Colon/metabolism , Colorectal Neoplasms/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Inhibitor of Differentiation Protein 1 , Intestinal Mucosa/metabolism , Mice , Mitotic Index , Oligonucleotides, Antisense/pharmacology , Precipitin Tests , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
2.
Mol Cell Biol ; 21(2): 524-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134340

ABSTRACT

The Id subfamily of helix-loop-helix (HLH) proteins plays a fundamental role in the regulation of cellular proliferation and differentiation. The major mechanism by which Id proteins are thought to inhibit differentiation is through interaction with other HLH proteins and inhibition of their DNA-binding activity. However, Id proteins have also been shown to interact with other proteins involved in regulating cellular proliferation and differentiation, suggesting a more widespread regulatory function. In this study we demonstrate functional interactions between Id proteins and members of the Pax-2/-5/-8 subfamily of paired-domain transcription factors. Members of the Pax transcription factor family have key functions in regulating several developmental processes exemplified by B lymphopoiesis, in which Pax-5 plays an essential role. Id proteins bind to Pax proteins in vitro and in vivo. Binding occurs through the paired DNA-binding domain of the Pax proteins and results in the disruption of DNA-bound complexes containing Pax-2, Pax-5, and Pax-8. In vivo, Id proteins modulate the transcriptional activity mediated by Pax-5 complexes on the B-cell-specific mb-1 promoter. Our results therefore demonstrate a novel facet of Id function in regulating cellular differentiation by functionally antagonizing the action of members of the Pax transcription factor family.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Helix-Loop-Helix Motifs , Neoplasm Proteins , Nuclear Proteins/antagonists & inhibitors , Repressor Proteins , Trans-Activators/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , 3T3 Cells , Animals , Antigens, CD/genetics , Base Sequence , CD79 Antigens , COS Cells , DNA/genetics , DNA/metabolism , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Inhibitor of Differentiation Proteins , Mice , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , PAX2 Transcription Factor , PAX5 Transcription Factor , PAX8 Transcription Factor , Paired Box Transcription Factors , Precipitin Tests , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, B-Cell/genetics , Trans-Activators/metabolism , ets-Domain Protein Elk-1
3.
EMBO J ; 18(4): 968-76, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-10022839

ABSTRACT

The Id subfamily of helix-loop-helix (HLH) proteins plays a fundamental role in the regulation of cellular proliferation and differentiation. Id proteins are thought to inhibit differentiation mainly through interaction with other HLH proteins and by blocking their DNA-binding activity. Members of the ternary complex factor (TCF) subfamily of ETS-domain proteins have key functions in regulating immediate-early gene expression in response to mitogenic stimulation. TCFs form DNA-bound complexes with the serum response factor (SRF) and are direct targets of MAP kinase (MAPK) signal transduction cascades. In this study we demonstrate functional interactions between Id proteins and TCFs. Ids bind to the ETS DNA-binding domain and disrupt the formation of DNA-bound complexes between TCFs and SRF on the c-fos serum response element (SRE). Inhibition occurs by disrupting protein-DNA interactions with the TCF component of this complex. In vivo, the Id proteins cause down-regulation of the transcriptional activity mediated by the TCFs and thereby block MAPK signalling to SREs. Therefore, our results demonstrate a novel facet of Id function in the coordination of mitogenic signalling and cell cycle entry.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Helix-Loop-Helix Motifs , Neoplasm Proteins , Repressor Proteins , Transcription Factors/metabolism , 3T3 Cells , Animals , DNA-Binding Proteins/genetics , Genes, fos/genetics , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Inhibitor of Differentiation Proteins , Mice , Nuclear Proteins/genetics , Oligodeoxyribonucleotides/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Serum Response Factor , Transcription Factors/genetics , ets-Domain Protein Elk-1 , ets-Domain Protein Elk-4
4.
Cell Growth Differ ; 9(12): 1015-24, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869302

ABSTRACT

Immunohistological detection of each of the four Id proteins (Id1-Id4) in sections of mouse testis revealed a unique temporal and spatial expression pattern for each Id protein during spermatogenesis. Furthermore, each Id protein displayed a distinctive, dynamic pattern of subcellular distribution. Id1 was uniquely expressed in MI/MII spermatocytes undergoing meiotic division. Id4 protein was detectable in the cytoplasm of type A1 spermatogonia, as well as in late pachytene and in diplotene spermatocytes. Id2 protein, which was most abundant in Sertoli cell nuclei, was also detectable in pachytene and diplotene spermatocytes, but as with Id4, it was absent from MI/MII cells. In postmeiotic spermatids, Id1, Id2, and Id4 proteins were expressed in a stage- and subcellular-specific manner. Expression of Id3 was restricted to Sertoli cell cytoplasm. In malignant seminoma cells, all four Id proteins were abundantly expressed with accompanying changes in their subcellular distribution. The observed expression of Id proteins in postproliferative Sertoli cells and spermatids and during specific stages of meiosis implies novel functional roles for this class of transcriptional regulator during spermatogenesis.


Subject(s)
Germ Cells/metabolism , Meiosis/genetics , Repressor Proteins , Sertoli Cells/metabolism , Spermatogenesis/genetics , Transcription Factors/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Humans , Immunohistochemistry , Inhibitor of Differentiation Protein 1 , Male , Mice , Mitosis/genetics , Seminoma , Spermatids/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/metabolism , Transcription Factors/metabolism
5.
Trends Cell Biol ; 8(2): 58-65, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9695810

ABSTRACT

Id helix-loop-helix proteins function at a general level as positive regulators of cell growth and as negative regulators of cell differentiation. They act as dominant-negative antagonists of other helix-loop-helix transcription factors, which drive cell lineage commitment and differentiation in diverse cell types of higher eukaryotes. In addition, the functions of Id proteins are integrated with cell-cycle-regulatory pathways orchestrated by cyclin-dependent kinases and the retinoblastoma protein. Here, some of the recent advances that highlight the importance of Id proteins as regulatory intermediates for coordinating differentiation-linked gene expression with cell-cycle control in response to extracellular signalling are reviewed.


Subject(s)
Eukaryotic Cells/physiology , Helix-Loop-Helix Motifs/genetics , Repressor Proteins , Transcription Factors/genetics , Animals , Cell Differentiation/physiology , Cell Division/physiology , Eukaryotic Cells/chemistry , Eukaryotic Cells/cytology , Inhibitor of Differentiation Protein 1 , Molecular Sequence Data , Sequence Homology, Amino Acid , Transcription Factors/metabolism
6.
J Biol Chem ; 273(14): 8278-86, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525934

ABSTRACT

Accumulating evidence implicates functions of the Id family of helix-loop-helix proteins in the regulation of cell growth and differentiation in metazoa. Within the mammalian hematopoietic organ, expression of the Id3 gene is restricted to the lymphoid cell compartment. We show here that in non-lymphoid hematopoietic cells, repression of transcription is correlated with hypermethylation of sequences in the vicinity of the upstream regulatory region of the Id3 gene, suggestive of a strict developmental control of expression of this gene in lymphoid versus non-lymphoid hematopoietic cells. Enforced ectopic expression of Id3 in K562 erythroid progenitor cells promotes erythroid differentiation and is correlated with a quantitative/qualitative shift in the profile of interacting TAL1 and E protein heterodimers that bind to a consensus E box sequence in in vitro band shift assays, consistent with selective targeting of E2A E protein(s) by Id3 and suggesting a possible mechanism involving TAL1-mediated differentiation. By using a Gal 4-VP16 two-hybrid competition assay and an E box-dependent reporter assay, we demonstrate directly that the E2A protein E47 preferentially associates with Id3 in vivo. These observations provide a paradigm for understanding how overlapping but distinct specificities of individual Id proteins may constitute a developmentally regulated program underlying cell determination in diverse lineages.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Leukemia, Erythroblastic, Acute/genetics , Neoplasm Proteins , Transcription Factors/genetics , Cell Differentiation/genetics , Cell Line , Helix-Loop-Helix Motifs , Hematopoietic Stem Cells/pathology , Humans , Inhibitor of Differentiation Proteins , Leukemia, Erythroblastic, Acute/pathology , Transcription Factors/biosynthesis , Transcription Factors/metabolism
7.
Mol Cell Biol ; 17(12): 6815-21, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9372912

ABSTRACT

The functions of basic helix-loop-helix (bHLH) transcription factors in activating differentiation-linked gene expression and in inducing G1 cell cycle arrest are negatively regulated by members of the Id family of HLH proteins. These bHLH antagonists are induced during a mitogenic signalling response, and they function by sequestering their bHLH targets in inactive heterodimers that are unable to bind to specific gene regulatory (E box) sequences. Recently, cyclin E-Cdk2- and cyclin A-Cdk2-dependent phosphorylation of a single conserved serine residue (Ser5) in Id2 has been shown to occur during late G1-to-S phase transition of the cell cycle, and this neutralizes the function of Id2 in abrogating E-box-dependent bHLH homo- or heterodimer complex formation in vitro (E. Hara, M. Hall, and G. Peters, EMBO J. 16:332-342, 1997). We now show that an analogous cell-cycle-regulated phosphorylation of Id3 alters the specificity of Id3 for abrogating both E-box-dependent bHLH homo- or heterodimer complex formation in vitro and E-box-dependent reporter gene function in vivo. Furthermore, compared with wild-type Id3, an Id3 Asp5 mutant (mimicking phosphorylation) is unable to promote cell cycle S phase entry in transfected fibroblasts, whereas an Id3 Ala5 mutant (ablating phosphorylation) displays an activity significantly greater than that of wild-type Id3 protein. Cdk2-dependent phosphorylation therefore provides a switch during late G1-to-S phase that both nullifies an early G1 cell cycle regulatory function of Id3 and modulates its target bHLH specificity. These data also demonstrate that the ability of Id3 to promote cell cycle S phase entry is not simply a function of its ability to modulate bHLH heterodimer-dependent gene expression and establish a biologically important mechanism through which Cdk2 and Id-bHLH functions are integrated in the coordination of cell proliferation and differentiation.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/physiology , Cyclin-Dependent Kinases/metabolism , Neoplasm Proteins , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Baculoviridae/genetics , Cell Line , Cyclin-Dependent Kinase 2 , Escherichia coli/genetics , Gene Expression , Helix-Loop-Helix Motifs/genetics , Helix-Loop-Helix Motifs/physiology , Humans , Inhibitor of Differentiation Proteins , Mutation , Phosphorylation , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera , Transcription Factors/genetics , Transfection
8.
J Biol Chem ; 271(39): 23603-6, 1996 Sep 27.
Article in English | MEDLINE | ID: mdl-8798572

ABSTRACT

Members of the Id family of helix-loop-helix proteins function as negative regulators of DNA binding, E protein, helix-loop-helix transcription factors in the control of cell growth, differentiation, and development. By using transient transfection analysis of COS cells, we show that in the absence of its E protein target, the Id3 protein is localized exclusively to the cytoplasm/perinuclear region. Co-transfection with E protein (E47) results in nuclear translocation of the Id3 protein, a process requiring both a functional Id helix-loop-helix dimerization domain and an E protein nuclear localization signal. Id3 that is associated with E protein displays an extended half-life, while the E protein itself is more rapidly turned over. These observations demonstrate that E protein, by nuclear chaperoning Id, can regulate the available cellular pool of its own inhibitory partner.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasm Proteins , Transcription Factors/metabolism , Animals , COS Cells , Cell Compartmentation , Cell Nucleus/metabolism , Helix-Loop-Helix Motifs , Humans , Inhibitor of Differentiation Proteins , Molecular Chaperones/metabolism , Precipitin Tests , Protein Binding , Recombinant Proteins , Structure-Activity Relationship , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transfection
9.
FEBS Lett ; 393(1): 113-6, 1996 Sep 09.
Article in English | MEDLINE | ID: mdl-8804437

ABSTRACT

The Id family of helix-loop-helix proteins function as negative regulators of DNA binding, basic helix-loop-helix proteins in the regulation of cell growth and differentiation. We report here on the identification of a 17 kDa variant of the 14 kDa Id-3 protein termed Id-3L (long version) which possesses a unique 60 amino acid carboxy-terminus generated by read through of a 'coding intron' and alternative splicing. Northern analysis revealed expression of a minor 1.1 kb Id-3L transcript together with the predominant 0.95 kb Id-3 transcript in the majority of adult human tissues analysed. The variant Id-3L protein is functionally distinguishable from conventional Id-3 since in in vitro DNA mobility shift assays, it was greatly impaired in its ability to abrogate binding of the basic helix-loop-helix protein, E47, to an E box recognition sequence.


Subject(s)
Alternative Splicing , Helix-Loop-Helix Motifs , Neoplasm Proteins , Transcription Factors/genetics , Adult , B-Lymphocytes/cytology , Blotting, Northern , Cells, Cultured , Gene Expression , Genetic Variation , Humans , Inhibitor of Differentiation Proteins , RNA/analysis , Transcription Factors/physiology
12.
Gene ; 151(1-2): 309-14, 1994 Dec 30.
Article in English | MEDLINE | ID: mdl-7828896

ABSTRACT

The helix-loop-helix (HLH) family of transcription factors plays a central role in the regulation of cell growth, differentiation and tumourigenesis. Members of the Id (inhibitor of DNA binding) class of these nuclear proteins are able to heterodimerise with and thereby antagonise the functions of other transcription factors of this family. We report here on the genomic organisation of the human Id3 (HLH 1R21/heir1) gene. Comparison with the two other mammalian Id genes, Id1 and Id2, reveals a highly conserved protein coding gene organisation consistent with evolution from a common, ancestral Id-like gene. In addition, by using a yeast artificial chromosome (YAC) clone of Id3, we have fine-scale mapped the gene to chromosome band 1p36.1 by fluorescence in situ hybridisation (FISH) and, using the same FISH technique, we have detected heterogeneity in tumour-associated 1p36 chromosome translocations.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 1 , Neoplasm Proteins , Transcription Factors/genetics , Adenocarcinoma/genetics , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Breast Neoplasms/genetics , Chromosome Mapping , Consensus Sequence , Conserved Sequence , Helix-Loop-Helix Motifs , Humans , In Situ Hybridization, Fluorescence , Inhibitor of Differentiation Proteins , Leukemia/genetics , Lung Neoplasms/genetics , Lymphocytes/cytology , Lymphocytes/physiology , Mammals , Molecular Sequence Data , Restriction Mapping , Sequence Homology, Amino Acid , Translocation, Genetic , Tumor Cells, Cultured
13.
Biochim Biophys Acta ; 1219(1): 160-2, 1994 Sep 13.
Article in English | MEDLINE | ID: mdl-8086456

ABSTRACT

We have determined the cDNA sequence encoding a 154 amino acid human Id-1 helix-loop-helix protein. Comparison with the amino acid sequences of human and mouse Id-2 and Id-3 proteins, reveals conservation/divergence of several residues in the helix-loop-helix domain known to be important for heterodimerisation, together with a common casein kinase II phosphorylation site.


Subject(s)
DNA-Binding Proteins/genetics , Helix-Loop-Helix Motifs , Repressor Proteins , Transcription Factors , Amino Acid Sequence , Base Sequence , Casein Kinase II , Conserved Sequence , DNA, Complementary/genetics , Humans , Inhibitor of Differentiation Protein 1 , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Sequence Homology, Amino Acid
14.
Biochim Biophys Acta ; 1216(2): 314-6, 1993 Nov 16.
Article in English | MEDLINE | ID: mdl-8241276

ABSTRACT

We have determined the cDNA sequence of a human B cell specific, immediate early gene, designated 1R20, which is inducible in response to several B cell activation signals. The cDNA sequence predicts a 196 amino acid open reading frame comprising numerous highly basic residues and the predicted structure contains several potential alpha helical domains together with eight consensus protein phosphorylation sites. The 1R20 gene has been localised by fluorescence in situ hybridisation to chromosome band 1q31, a region known to be implicated in the pathogenesis of haemopoietic malignancies.


Subject(s)
B-Lymphocytes/ultrastructure , Chromosomes, Human, Pair 1 , Genes, Immediate-Early , Immediate-Early Proteins/genetics , Phosphoproteins/genetics , RGS Proteins , Amino Acid Sequence , B-Lymphocytes/drug effects , Base Sequence , Binding Sites , Conserved Sequence , DNA, Complementary/chemistry , Humans , Immediate-Early Proteins/chemistry , Lymphoma, B-Cell/genetics , Molecular Sequence Data , Phosphoproteins/chemistry
15.
Oncogene ; 8(3): 599-607, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8437843

ABSTRACT

Transcription factors characterized by the presence of a helix-loop-helix (HLH) domain play a central role in the regulation of cell growth/differentiation and tumorigenesis. We report here the cDNA sequence of a human early-response gene, designated HLH 1R21, encoding a 15-kDa HLH protein that lacks a basic, DNA-binding domain and which by a number of criteria appears to be the human homologue of mouse HLH 462. Like its murine counterpart, HLH 1R21 protein functions as an Id (inhibitor of DNA binding) transcription factor by inhibiting the binding of E2A-containing protein complexes to muscle creatine kinase E-box enhancer oligonucleotide in vitro. However HLH 1R21 does not inhibit the binding of HLH Max protein to a Max-binding oligonucleotide in vitro, indicating that it has limited promiscuity in its ability to antagonize the function of other HLH transcription factors. In addition, HLH 1R21 mRNA transcripts are regulated by phorbol ester treatment of a diverse range of human cell lines and, when overexpressed in mouse NIH3T3 cells, HLH 1R21 induces a morphologically transformed phenotype.


Subject(s)
DNA-Binding Proteins/genetics , Genome, Human , Immunoglobulin Idiotypes/genetics , Protein Kinase C/physiology , Transcription Factors/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Transformation, Neoplastic , DNA-Binding Proteins/chemistry , Enzyme Activation , Gene Expression Regulation/drug effects , Humans , Inhibitor of Differentiation Proteins , Mice , Molecular Sequence Data , Proteins/chemistry , Sequence Homology, Amino Acid , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...