201
|
Ordentlich P, Downes M, Xie W, Genin A, Spinner NB, Evans RM. Unique forms of human and mouse nuclear receptor corepressor SMRT. Proc Natl Acad Sci U S A 1999; 96:2639-44. [PMID: 10077563 PMCID: PMC15821 DOI: 10.1073/pnas.96.6.2639] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nuclear hormone receptors have been shown to repress transcription in the absence of ligand. This repression is mediated by a corepressor complex that contains the Sin3A protein and histone deacetylases (HDAC1 and 2). Studies by several groups demonstrate that this complex is recruited to nuclear receptors through the highly related corepressors SMRT (silencing mediator of retinoid acid and thyroid hormone receptor) and N-CoR (nuclear receptor corepressor). We describe here the cloning, characterization, and chromosomal mapping of forms of human and mouse SMRT that includes a 1,000-aa extension, which reveals striking homology to the amino terminus of N-CoR. Structure and function studies of wild-type and natural splicing variants suggest the presence of 3-4 amino terminal domains that repress in a cooperative as well as mechanistically distinct fashion.
Collapse
Affiliation(s)
- P Ordentlich
- Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
202
|
Dang VD, Benedik MJ, Ekwall K, Choi J, Allshire RC, Levin HL. A new member of the Sin3 family of corepressors is essential for cell viability and required for retroelement propagation in fission yeast. Mol Cell Biol 1999; 19:2351-65. [PMID: 10022921 PMCID: PMC84027 DOI: 10.1128/mcb.19.3.2351] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tf1 is a long terminal repeat (LTR)-containing retrotransposon that propagates within the fission yeast Schizosaccharomyces pombe. LTR-retrotransposons possess significant similarity to retroviruses and therefore serve as retrovirus models. To determine what features of the host cell are important for the proliferation of this class of retroelements, we screened for mutations in host genes that reduced the transposition activity of Tf1. We report here the isolation and characterization of pst1(+), a gene required for Tf1 transposition. The predicted amino acid sequence of Pst1p possessed high sequence homology with the Sin3 family of proteins, known for their interaction with histone deacetylases. However, unlike the SIN3 gene of Saccharomyces cerevisiae, pst1(+) is essential for cell viability. Immunofluorescence microscopy indicated that Pst1p was localized in the nucleus. Consistent with the critical role previously reported for Sin3 proteins in the histone acetylation process, we found that the growth of the strain with the pst1-1 allele was supersensitive to the specific histone deacetylase inhibitor trichostatin A. However, our analysis of strains with the pst1-1 mutation was unable to detect any changes in the acetylation of specific lysines of histones H3 and H4 as measured in bulk chromatin. Interestingly, the pst1-1 mutant strain produced wild-type levels of Tf1-encoded proteins and cDNA, indicating that the defect in transposition occurred after reverse transcription. The results of immunofluorescence microscopy showed that the nuclear localization of the Tf1 capsid protein was disrupted in the strain with the pst1-1 mutation, indicating an important role of pst1(+) in modulating the nuclear import of Tf1 virus-like particles.
Collapse
Affiliation(s)
- V D Dang
- Laboratory of Eukaryotic Gene Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
203
|
Greenberg RA, O'Hagan RC, Deng H, Xiao Q, Hann SR, Adams RR, Lichtsteiner S, Chin L, Morin GB, DePinho RA. Telomerase reverse transcriptase gene is a direct target of c-Myc but is not functionally equivalent in cellular transformation. Oncogene 1999; 18:1219-26. [PMID: 10022128 DOI: 10.1038/sj.onc.1202669] [Citation(s) in RCA: 295] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The telomerase reverse transcriptase component (TERT) is not expressed in most primary somatic human cells and tissues, but is upregulated in the majority of immortalized cell lines and tumors. Here, we identify the c-Myc transcription factor as a direct mediator of telomerase activation in primary human fibroblasts through its ability to specifically induce TERT gene expression. Through the use of a hormone inducible form of c-Myc (c-Myc-ER), we demonstrate that Myc-induced activation of the hTERT promoter requires an evolutionarily conserved E-box and that c-Myc-ER-induced accumulation of hTERT mRNA takes place in the absence of de novo protein synthesis. These findings demonstrate that the TERT gene is a direct transcriptional target of c-Myc. Since telomerase activation frequently correlates with immortalization and telomerase functions to stabilize telomers in cycling cells, we tested whether Myc-induced activation of TERT gene expression represents an important mechanism through which c-Myc acts to immortalize cells. Employing the rat embryo fibroblast cooperation assay, we show that TERT is unable to substitute for c-Myc in the transformation of primary rodent fibroblasts, suggesting that the transforming activities of Myc extend beyond its ability to activate TERT gene expression and hence telomerase activity.
Collapse
Affiliation(s)
- R A Greenberg
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Lee TC, Ziff EB. Mxi1 is a repressor of the c-Myc promoter and reverses activation by USF. J Biol Chem 1999; 274:595-606. [PMID: 9872993 DOI: 10.1074/jbc.274.2.595] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The basic region/helix-loop-helix/leucine zipper (B-HLH-LZ) oncoprotein c-Myc is abundant in proliferating cells and forms heterodimers with Max protein that bind to E-box sites in DNA and stimulate genes required for proliferation. A second B-HLH-LZ protein, Mxi1, is induced during terminal differentiation, and forms heterodimers with Max that also bind E-boxes but tether the mSin3 transcriptional repressor protein along with histone deacetylase thereby antagonizing Myc-dependent activation. We show that Mxi1 also antagonizes Myc by a second pathway, repression of transcription from the major c-myc promoter, P2. Repression was independent of Mxi1 binding to mSin3 but dependent on the Mxi1 LZ and COOH-terminal sequences, including putative casein kinase II phosphorylation sites. Repression targeted elements of the myc P2 promoter core (-35/+10), where it reversed transactivation by the constitutive transcription factor, USF. We show that Zn2+ induction of a stably transfected, metallothionein promoter-regulated mxi1 gene blocked the ability of serum to induce transcription of the endogenous c-myc gene and cell entry into S phase. Thus, induction of Mxi1 in terminally differentiating cells may block Myc function by repressing the c-myc gene P2 promoter, as well as by antagonizing Myc-dependent transactivation through E-boxes.
Collapse
Affiliation(s)
- T C Lee
- Howard Hughes Medical Institute, Department of Biochemistry and Kaplan Cancer Center, New York University Medical Center, New York, New York 10016, USA
| | | |
Collapse
|
205
|
Affiliation(s)
- C V Dang
- Department of Medicine, The Johns Hopkins Oncology Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
206
|
Torres R, Firestein BL, Dong H, Staudinger J, Olson EN, Huganir RL, Bredt DS, Gale NW, Yancopoulos GD. PDZ proteins bind, cluster, and synaptically colocalize with Eph receptors and their ephrin ligands. Neuron 1998; 21:1453-63. [PMID: 9883737 DOI: 10.1016/s0896-6273(00)80663-7] [Citation(s) in RCA: 362] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Localizing cell surface receptors to specific subcellular positions can be critical for their proper functioning, as most notably demonstrated at neuronal synapses. PDZ proteins apparently play critical roles in such protein localizations. Receptor tyrosine kinases have not been previously shown to interact with PDZ proteins in vertebrates. We report that Eph receptors and their membrane-linked ligands all contain PDZ recognition motifs and can bind and be clustered by PDZ proteins. In addition, we find that Eph receptors and ligands colocalize with PDZ proteins at synapses. Thus, PDZ proteins may play critical roles in localizing vertebrate receptor tyrosine kinases and/or their ligands and may be particularly important for Eph function in guidance or patterning or at the synapse.
Collapse
Affiliation(s)
- Richard Torres
- Regeneron Pharmaceuticals, Tarrytown, New York 10591-6707, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Lutterbach B, Westendorf JJ, Linggi B, Patten A, Moniwa M, Davie JR, Huynh KD, Bardwell VJ, Lavinsky RM, Rosenfeld MG, Glass C, Seto E, Hiebert SW. ETO, a target of t(8;21) in acute leukemia, interacts with the N-CoR and mSin3 corepressors. Mol Cell Biol 1998; 18:7176-84. [PMID: 9819404 PMCID: PMC109299 DOI: 10.1128/mcb.18.12.7176] [Citation(s) in RCA: 362] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/1998] [Accepted: 08/27/1998] [Indexed: 11/20/2022] Open
Abstract
t(8;21) is one of the most frequent translocations associated with acute myeloid leukemia. It produces a chimeric protein, acute myeloid leukemia-1 (AML-1)-eight-twenty-one (ETO), that contains the amino-terminal DNA binding domain of the AML-1 transcriptional regulator fused to nearly all of ETO. Here we demonstrate that ETO interacts with the nuclear receptor corepressor N-CoR, the mSin3 corepressors, and histone deacetylases. Endogenous ETO also cosediments on sucrose gradients with mSin3A, N-CoR, and histone deacetylases, suggesting that it is a component of one or more corepressor complexes. Deletion mutagenesis indicates that ETO interacts with mSin3A independently of its association with N-CoR. Single amino acid mutations that impair the ability of ETO to interact with the central portion of N-CoR affect the ability of the t(8;21) fusion protein to repress transcription. Finally, AML-1/ETO associates with histone deacetylase activity and a histone deacetylase inhibitor impairs the ability of the fusion protein to repress transcription. Thus, t(8;21) fuses a component of a corepressor complex to AML-1 to repress transcription.
Collapse
Affiliation(s)
- B Lutterbach
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Warrell RP, He LZ, Richon V, Calleja E, Pandolfi PP. Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 1998; 90:1621-5. [PMID: 9811311 DOI: 10.1093/jnci/90.21.1621] [Citation(s) in RCA: 393] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Acetylation of DNA-associated histones is linked to activation of gene transcription, whereas histone deacetylation is associated with transcriptional repression. Recent studies have shown that inhibitors of histone deacetylases can relieve transcriptional repression caused by the products of certain oncogenes. We tested whether these findings could be applied clinically to a patient with highly resistant acute promyelocytic leukemia. METHODS A patient who had experienced multiple relapses was treated with all-trans-retinoic acid alone and in combination with sodium phenylbutyrate, an inhibitor of histone deacetylases. Immunohistochemistry and western blot analysis were used to assay for histone hyperacetylation in mononuclear cells from the patient's blood and bone marrow. Marrow mononuclear cells and reverse transcription-polymerase chain reaction (RT-PCR) analysis of messenger RNA encoded by the PML/RAR-alpha oncogene were used to assess minimal residual disease. RESULTS The patient proved clinically resistant to treatment with all-trans-retinoic acid alone. However, 23 days after sodium phenylbutyrate was added to the treatment regimen, visible leukemic cells had been eliminated from her bone marrow, and she achieved a complete clinical and cytogenetic remission shortly thereafter. With a second treatment course, analysis for minimal residual disease by RT-PCR proved negative. Immunofluorescence and western blot analysis showed that phenylbutyrate caused a time-dependent increase in histone acetylation in blood and bone marrow mononuclear cells. CONCLUSIONS Clinical treatment with an inhibitor of histone deacetylase induces histone hyperacetylation in target cells and may restore sensitivity to the anti-leukemic effects of all-trans-retinoic acid in acute promyelocytic leukemia. Similar therapy may prove useful in other neoplastic diseases that are associated with oncogenic repression of gene transcription due to recruitment of histone deacetylases.
Collapse
Affiliation(s)
- R P Warrell
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, and the Cornell University Medical College, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
209
|
Wong CW, Privalsky ML. Components of the SMRT corepressor complex exhibit distinctive interactions with the POZ domain oncoproteins PLZF, PLZF-RARalpha, and BCL-6. J Biol Chem 1998; 273:27695-702. [PMID: 9765306 DOI: 10.1074/jbc.273.42.27695] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many transcription factors function by repressing gene transcription. For a variety of these transcription factors the ability to physically recruit auxiliary proteins, denoted corepressors, is crucial for the ability to silence gene expression. We and others have previously implicated the SMRT corepressor in the actions of the PLZF transcription factor and in the function of its oncogenic derivative, PLZF-retinoic acid receptor (RARalpha), in promyelocytic leukemia. We report here that PLZF, and a structurally similar transcriptional repressor, BCL-6, can interact with a variety of corepressor proteins in addition to SMRT, including the mSin3A protein and (for PLZF) histone deacetylase-1. Unexpectedly, these additional interactions with corepressor components are nonequivalent for these otherwise similar oncoproteins, suggesting that transcriptional repression by BCL-6 and by PLZF may differ in mechanism. Furthermore, we demonstrate that the oncogenic PLZF-RARalpha chimera lacks several important corepressor interaction sites that are present in the native PLZF protein. Thus the t(11;17) translocation that creates the PLZF-RARalpha chimera generates an oncoprotein with potentially novel regulatory properties distinct from those of either parental protein. Our results demonstrate that otherwise similar transcription factors can differ notably in their interactions with the corepressor machinery.
Collapse
Affiliation(s)
- C W Wong
- Section of Microbiology, Division of Biological Sciences, University of California, Davis, California 95616, USA
| | | |
Collapse
|
210
|
Workman JL, Kingston RE. Alteration of nucleosome structure as a mechanism of transcriptional regulation. Annu Rev Biochem 1998; 67:545-79. [PMID: 9759497 DOI: 10.1146/annurev.biochem.67.1.545] [Citation(s) in RCA: 882] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The nucleosome, which is the primary building block of chromatin, is not a static structure: It can adopt alternative conformations. Changes in solution conditions or changes in histone acetylation state cause nucleosomes and nucleosomal arrays to behave with altered biophysical properties. Distinct subpopulations of nucleosomes isolated from cells have chromatographic properties and nuclease sensitivity different from those of bulk nucleosomes. Recently, proteins that were initially identified as necessary for transcriptional regulation have been shown to alter nucleosomal structure. These proteins are found in three types of multiprotein complexes that can acetylate nucleosomes, deacetylate nucleosomes, or alter nucleosome structure in an ATP-dependent manner. The direct modification of nucleosome structure by these complexes is likely to play a central role in appropriate regulation of eukaryotic genes.
Collapse
Affiliation(s)
- J L Workman
- Howard Hughes Medical Institute, Pennsylvania State University, University Park 16802, USA.
| | | |
Collapse
|
211
|
Wong CW, Privalsky ML. Transcriptional repression by the SMRT-mSin3 corepressor: multiple interactions, multiple mechanisms, and a potential role for TFIIB. Mol Cell Biol 1998; 18:5500-10. [PMID: 9710634 PMCID: PMC109135 DOI: 10.1128/mcb.18.9.5500] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/1998] [Accepted: 06/11/1998] [Indexed: 11/20/2022] Open
Abstract
A variety of eukaryotic transcription factors, including the nuclear hormone receptors, Max-Mad, BCL-6, and PLZF, appear to mediate transcriptional repression through the ability to recruit a multiprotein corepressor complex to the target promoter. This corepressor complex includes the SMRT/N-CoR polypeptides, mSin3A or -B, and histone deacetylase 1 or 2. The presence of a histone-modifying activity in the corepressor complex has led to the suggestion that gene silencing is mediated by modification of the chromatin template, perhaps rendering it less accessible to the transcriptional machinery. We report here, however, that the corepressor complex actually appears to exhibit multiple mechanisms of transcriptional repression, only one of which corresponds with detectable recruitment of the histone deacetylase. We provide evidence instead of an alternative pathway of repression that may be mediated by direct physical interactions between components of the corepressor complex and the general transcription factor TFIIB.
Collapse
Affiliation(s)
- C W Wong
- Section of Microbiology, Division of Biological Sciences, University of California at Davis, Davis, California 95616, USA
| | | |
Collapse
|
212
|
Braun K, Hölzl G, Pusch O, Hengstschläger M. Deregulated expression of CDK2- or CDK3-associated kinase activities enhances c-Myc-induced apoptosis. DNA Cell Biol 1998; 17:789-98. [PMID: 9778038 DOI: 10.1089/dna.1998.17.789] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Activation of high ectopic levels of c-Myc in serum-deprived Rat1-MycER cells by 4-hydroxytamoxifen induces both proliferation and apoptosis. To further elucidate the role of G1 cyclin-dependent kinases (CDKs) in the process of Myc-induced apoptosis, we generated Rat1-MycER cells stably overexpressing CDK2 or CDK3. Ectopic expression of these CDKs in Myc-overexpressing cells was accompanied by upregulation of the specific kinase activities. Whereas neither high ectopic CDK2 nor CDK3 alone induced apoptosis in serum-deprived Rat1 cells, both CDKs markedly elevated the incidence of Myc-induced apoptosis. It was shown earlier that in Rat1-MycER cells, which are resistant to tumor necrosis factor-alpha (TNF) when grown in high serum concentrations, the addition of TNF with the concomitant activation of Myc resulted in apoptotic cell death. Here, we show that neither CDK2 nor CDK3 induces susceptibility to the cytotoxic action of TNF in Rat1 cells. However, both molecules heavily elevated the incidence of apoptosis induced by TNF together with Myc. It has earlier been reported that Myc-induced apoptosis in serum-deprived Rat1 fibroblasts is inhibited by specific cytokines, such as platelet-derived growth factor (PDGF). Here, we demonstrate that PDGF-mediated protection from Myc-induced apoptosis is almost lost in Rat1 cells overexpressing CDK2 or CDK3. These apoptotic effects of CDK2 or CDK3 are not accompanied by alterations of proliferation parameters, such as DNA distribution, time the cells spend in each phase of the cell cycle, thymidine incorporation into DNA, or cell size analyzed during Myc-induced apoptosis. However, we found CDK3 to deregulate E2F-dependent transcription. In this report, we provide evidence for a not yet described property of CDK2 or CDK3 besides their activity in promoting proliferation: these G1-CDKs can promote apoptosis by interfering with the cell's response to survival factors.
Collapse
Affiliation(s)
- K Braun
- Obstetrics and Gynecology, Department of Prenatal Diagnosis and Therapy, University of Vienna, Austria
| | | | | | | |
Collapse
|
213
|
McMahon SB, Van Buskirk HA, Dugan KA, Copeland TD, Cole MD. The novel ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F oncoproteins. Cell 1998; 94:363-74. [PMID: 9708738 DOI: 10.1016/s0092-8674(00)81479-8] [Citation(s) in RCA: 508] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The c-Myc and E2F transcription factors are among the most potent regulators of cell cycle progression in higher eukaryotes. This report describes the isolation of a novel, highly conserved 434 kDa protein, designated TRRAP, which interacts specifically with the c-Myc N terminus and has homology to the ATM/PI3-kinase family. TRRAP also interacts specifically with the E2F-1 transactivation domain. Expression of transdominant mutants of the TRRAP protein or antisense RNA blocks c-Myc- and E1A-mediated oncogenic transformation. These data suggest that TRRAP is an essential cofactor for both the c-Myc and E1A/E2F oncogenic transcription factor pathways.
Collapse
Affiliation(s)
- S B McMahon
- Department of Molecular Biology, Princeton University, New Jersey 08544-1014, USA
| | | | | | | | | |
Collapse
|
214
|
Pei L. Molecular cloning of a novel transcriptional repressor protein of the rat type 1 vasoactive intestinal peptide receptor gene. J Biol Chem 1998; 273:19902-8. [PMID: 9677428 DOI: 10.1074/jbc.273.31.19902] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
This study demonstrates that the transcriptional repressor sequence of the rat vasoactive intestinal peptide receptor (VIPR) gene constitutes a 42-base pair core element that is the binding site for a nuclear protein. We showed that this element was able to confer transcriptional repression to a heterologous promoter and that deletion or point mutations within this element resulted in loss of transcriptional repression. Southwestern blot analysis indicated that the VIPR repressor element interacts specifically with a nuclear protein of about 72 kDa. By screening a rat lung expression library coupled with rapid amplification of cDNA ends polymerase chain reactions, we isolated a cDNA clone (designated as VIPR-RP) that contains an open reading frame of 656 amino acids. VIPR-RP is 78% identical to a previously characterized protein, differentiation-specific element-binding protein, which is a member of a family of proteins including components of the DNA replication factor C complex. However, VIPR-RP cDNA encodes for a much smaller protein than differentiation-specific element-binding protein because of a frameshift. VIPR-RP mRNA is expressed in multiple tissues, including lung, liver, brain, heart, kidney, spleen, and testis. VIPR-RP protein specifically interacts with the VIPR repressor element as demonstrated by gel shift assays. Transfection of VIP-RP expression vector into Cos cells resulted in transcriptional repression of a reporter construct containing multiple copies of the VIPR repressor element. These results indicate that VIPR-RP is a novel transcriptional repressor protein that regulates VIPR expression.
Collapse
Affiliation(s)
- L Pei
- Division of Endocrinology, Cedars-Sinai Research Institute, UCLA School of Medicine, Los Angeles, California 90048, USA
| |
Collapse
|
215
|
Abstract
Taken together, the available data appear to be consistent with a model in which Myc proteins function downstream of D-type cyclins and synergize with E2F proteins in the activation of the cyclin E/cdk2 kinase. This view of Myc proteins appears strikingly similar to established models for the E2F/DP family of proteins. However, it should be noted that there are clear differences and several predictions of such a model that have been critically tested for E2F proteins are still untested for Myc in this model. First, it appears that at least some target genes of Myc implicated in this process are still unknown; second, clear data from knockout cells that link p107 to Myc function are missing; and third, we are not aware of studies of tumour samples that clarify whether mutations in myc genes relieve the requirement for mutations in the cyclin D/p16 pathway.
Collapse
Affiliation(s)
- A Bürgin
- Institut für Molekularbiologie und Tumorforschung (IMT), Universität Marburg, Germany
| | | | | |
Collapse
|
216
|
Hua SB, Luo Y, Qiu M, Chan E, Zhou H, Zhu L. Construction of a modular yeast two-hybrid cDNA library from human EST clones for the human genome protein linkage map. Gene 1998; 215:143-52. [PMID: 9666106 DOI: 10.1016/s0378-1119(98)00258-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Identification of all human protein-protein interactions will lead to a global human protein linkage map that will provide important information for functional genomics studies. The yeast two-hybrid system is a powerful molecular genetic approach for studying protein-protein interactions. To apply this technology to generate a human protein linkage map, the first step is to construct two-hybrid cDNA libraries that cover the entire human genome. With a homologous recombination-mediated approach, we have constructed a modular human EST-derived yeast two-hybrid library in the Gal4 activation domain-based vector, pACT2. Quality analysis of this library indicated that the approach of constructing two-hybrid cDNA libraries from individually arrayed human EST clones is feasible, and such a two-hybrid library is suitable for detecting protein-protein interactions. This is also the first time that a comprehensive two-hybrid system cDNA library has been constructed from a collection of individually arrayed EST clones.
Collapse
Affiliation(s)
- S B Hua
- Gene, Net Group, CLONTECH Laboratories Inc., 1020 East Meadow Circle, Palo Alto, CA 94303, USA.
| | | | | | | | | | | |
Collapse
|
217
|
Johnstone RW, Kerry JA, Trapani JA. The human interferon-inducible protein, IFI 16, is a repressor of transcription. J Biol Chem 1998; 273:17172-7. [PMID: 9642285 DOI: 10.1074/jbc.273.27.17172] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
IFI 16 is a member of a family of interferon-inducible proteins, including the human MNDA (myeloid nuclear differentiation antigen), the recently identified AIM-2 (absent in melanoma), and the homologous murine molecules, p202, p204, and D3. IFI 16 contains a domain at the amino terminus capable of binding double-stranded DNA and a bipartite nuclear localization signal. No molecular or biological function has been assigned to any of the human family members, although a role in transcription regulation has been proposed. In the present study, we show IFI 16 fused to the GAL4 DNA binding domain can function as a transcriptional repressor. IFI 16-mediated repression is not dependent on the position or distance of IFI 16 binding, relative to the site of transcription initiation, and it can significantly repress when only one GAL4 DNA element is present in the promoter. We mapped the transcriptional repression domains to the 200 amino acid repeat regions common to all human and mouse family members. We also demonstrate that wild type IFI 16 can repress transcription of a reporter gene containing the minimal promoter region of the human cytomegalovirus UL54 gene. Thus, IFI 16 is a transcriptional repressor, with a modular structure typical of many known transcription regulators.
Collapse
Affiliation(s)
- R W Johnstone
- The Austin Research Institute, Austin Hospital, Studley Road, Heidelberg 3084, Victoria, Australia.
| | | | | |
Collapse
|
218
|
Relationship between histone acetylation/deacetylation and gene transcription. CHINESE SCIENCE BULLETIN-CHINESE 1998. [DOI: 10.1007/bf02883072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
219
|
Laherty CD, Billin AN, Lavinsky RM, Yochum GS, Bush AC, Sun JM, Mullen TM, Davie JR, Rose DW, Glass CK, Rosenfeld MG, Ayer DE, Eisenman RN. SAP30, a component of the mSin3 corepressor complex involved in N-CoR-mediated repression by specific transcription factors. Mol Cell 1998; 2:33-42. [PMID: 9702189 DOI: 10.1016/s1097-2765(00)80111-2] [Citation(s) in RCA: 167] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The transcriptional corepressor mSin3 is found in a large multiprotein complex containing the histone deacetylases HDAC1 and HDAC2, in addition to at least five tightly associated polypeptides. We have cloned and characterized a novel component of the mSin3 complex, SAP30, SAP30 binds to mSin3 and is capable of mediating transcriptional repression via histone deacetylases. SAP30 also binds the N-CoR corepressor and is required for N-CoR-mediated repression by antagonist-bound estrogen receptor and the homeodomain protein Rpx, as well as N-CoR suppression of transactivation by the POU domain protein Pit-1. However, SAP30 is not required for N-CoR-mediated repression by unliganded retinoic acid receptor or thyroid hormone receptor, suggesting that SAP30 is involved in the functional recruitment of the mSin3-histone deacetylase complex to a specific subset of N-CoR corepressor complexes.
Collapse
Affiliation(s)
- C D Laherty
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Schreiber-Agus N, Meng Y, Hoang T, Hou H, Chen K, Greenberg R, Cordon-Cardo C, Lee HW, DePinho RA. Role of Mxi1 in ageing organ systems and the regulation of normal and neoplastic growth. Nature 1998; 393:483-7. [PMID: 9624006 DOI: 10.1038/31008] [Citation(s) in RCA: 163] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mxi1 belongs to the Mad (Mxi1) family of proteins, which function as potent antagonists of Myc oncoproteins. This antagonism relates partly to their ability to compete with Myc for the protein Max and for consensus DNA binding sites and to recruit transcriptional co-repressors. Mad(Mxi1) proteins have been suggested to be essential in cellular growth control and/or in the induction and maintenance of the differentiated state. Consistent with these roles, mxi1 may be the tumour-suppressor gene that resides at region 24-26 of the long arm of chromosome 10. This region is a cancer hotspot, and mutations here may be involved in several cancers, including prostate adenocarcinoma. Here we show that mice lacking Mxi1 exhibit progressive, multisystem abnormalities. These mice also show increased susceptibility to tumorigenesis either following carcinogen treatment or when also deficient in Ink4a. This cancer-prone phenotype may correlate with the enhanced ability of several mxi1-deficient cell types, including prostatic epithelium, to proliferate. Our results show that Mxi1 is involved in the homeostasis of differentiated organ systems, acts as a tumour suppressor in vivo, and engages the Myc network in a functionally relevant manner.
Collapse
Affiliation(s)
- N Schreiber-Agus
- Department of Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Torchia J, Glass C, Rosenfeld MG. Co-activators and co-repressors in the integration of transcriptional responses. Curr Opin Cell Biol 1998; 10:373-83. [PMID: 9640539 DOI: 10.1016/s0955-0674(98)80014-8] [Citation(s) in RCA: 438] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The nuclear hormone receptors are DNA binding transcription factors that are regulated by binding of ligands, switching them from an inactive or repressive state to gene-activating functions. Recent evidence supports the hypothesis that many nuclear receptors switch, in a ligand-dependent manner, between binding of a multicomponent co-repressor complex containing histone deacetyltransferase activity, and binding of a co-activator complex containing factors with histone acetyltransferase activity that are further regulated by diverse signal transduction pathways. The identification of these limiting co-repressor and co-activator complexes and their specific interaction motifs, in concert with solution of the structures of the receptor ligand-binding domain in apo (empty) and ligand bound forms, indicates a common molecular mechanism by which these factors activate and repress gene transcription.
Collapse
Affiliation(s)
- J Torchia
- Howard Hughes Medical Institute, University of California San Diego 92093, USA.
| | | | | |
Collapse
|
222
|
Abstract
The mechanisms underlying transcriptional activation and repression have become much clearer. Recent evidence suggests that transcription factors that do not bind DNA directly, the co-activators and co-repressors, mediate a large number of cell signaling events. Their association with histone acetylases, to mediate activation, or deacetylases, to mediate repression, provide a model for explaining how gene expression is regulated.
Collapse
Affiliation(s)
- R H Goodman
- Vollum Institute, Oregon Health Sciences University, Portland 97201, USA.
| | | |
Collapse
|
223
|
Affiliation(s)
- M F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital Memphis, Tennessee 38105, USA
| |
Collapse
|
224
|
Walhout AJ, van der Vliet PC, Timmers HT. Sequences flanking the E-box contribute to cooperative binding by c-Myc/Max heterodimers to adjacent binding sites. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1397:189-201. [PMID: 9565685 DOI: 10.1016/s0167-4781(97)00227-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Previously, we have shown that c-Myc/Max heterodimers, bind cooperatively to the two adjacent, canonical E-boxes (CACGTG) located in the rat ornithine decarboxylase (ODC) gene. In order to study this in more detail, we changed the length of the linker that separates the two E-boxes, as well as their flanking sequences. We found that high affinity, cooperative binding requires a minimal linker length of 1-4 bp and that the binding affinity is influenced by E-box flanking sequences. Binding to the c-Myc responsive element of prothymosin alpha, containing both a canonical and a noncanonical E-box (CAAGTG) was also studied. As shown by DNAseI footprinting analysis, only the canonical E-box is bound by c-Myc/Max and c-Max/Max dimers. Replacing the noncanonical site with a canonical E-box only partially restored high affinity, cooperative binding. By making hybrid fragments between ODC and prothymosin alpha, we found that nucleotides in the linker between the E-boxes influence the affinity of c-Myc/Max heterodimers. Taken together, our results show that E-box sequences and sequences in the linker separating both E-boxes influence cooperative, high affinity binding by c-Myc/Max dimers.
Collapse
Affiliation(s)
- A J Walhout
- Laboratory for Physiological Chemistry, Utrecht University, PO Box 80042, 3508 TA Utrecht, Netherlands
| | | | | |
Collapse
|
225
|
Steiner P, Rudolph B, Müller D, Eilers M. The functions of Myc in cell cycle progression and apoptosis. PROGRESS IN CELL CYCLE RESEARCH 1998; 2:73-82. [PMID: 9552384 DOI: 10.1007/978-1-4615-5873-6_7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
c-myc has emerged as one of the central regulators of mammalian cell proliferation. The gene encodes a transcription factor of the HLH/leucine zipper family of proteins that activates transcription as part of a heteromeric complex with a protein termed Max. In mammalian fibroblasts, Myc acts as an upstream regulator of cyclin-dependent kinases and functionally antagonises the action of at least one cdk inhibitor, p27. Myc also induces cells to undergo apoptosis, and the relationship between Myc-induced cell cycle entry and apoptosis is discussed.
Collapse
Affiliation(s)
- P Steiner
- Zentrum für Molekulare Biologie Heidelberg (ZMBH)
| | | | | | | |
Collapse
|
226
|
Lo Nigro C, Venesio T, Reymond A, Meroni G, Alberici P, Cainarca S, Enrico F, Stack M, Ledbetter DH, Liscia DS, Ballabio A, Carrozzo R. The human ROX gene: genomic structure and mutation analysis in human breast tumors. Genomics 1998; 49:275-82. [PMID: 9598315 DOI: 10.1006/geno.1998.5241] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have recently isolated a human gene, ROX, encoding a new member of the basic helix-loop-helix leucine zipper protein family. ROX is capable of heterodimerizing with Max and acts as a transcriptional repressor in an E-box-driven reporter gene system, while it was found to activate transcription in HeLa cells. ROX expression levels vary during the cell cycle, being down-regulated in proliferating cells. These biological properties of ROX suggest a possible involvement of this gene in cell proliferation and differentiation. The ROX gene maps to chromosome 17p13.3, a region frequently deleted in human malignancies. Here we report the genomic structure of the human ROX gene, which is composed of six exons and spans a genomic region of less than 40 kb. In an attempt to identify possible inactivating mutations in the ROX gene in human breast cancer, we performed a single-strand conformation polymorphism analysis of its coding region in 16 sporadic breast carcinomas showing loss of heterozygosity in the 17p13.3 region. No mutations were found in this analysis. Five nucleotide polymorphisms were identified in the ROX gene, three of which caused an amino acid substitution. These nucleotide changes were present in the peripheral blood DNAs of both the patients and the control individuals. In vitro translated assays did not show a significant decrease in the ability of the ROX mutant proteins to bind DNA or to repress transcription of a driven reporter gene in HEK293 cells. Despite experimental evidence that ROX might act as a tumor suppressor gene, our data suggest that mutations in the coding region of ROX are uncommon in human breast tumorigenesis.
Collapse
Affiliation(s)
- C Lo Nigro
- Telethon Institute of Genetics and Medicine (TIGEM), San Raffaele Biomedical Science Park, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Pomerantz J, Schreiber-Agus N, Liégeois NJ, Silverman A, Alland L, Chin L, Potes J, Chen K, Orlow I, Lee HW, Cordon-Cardo C, DePinho RA. The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2's inhibition of p53. Cell 1998; 92:713-23. [PMID: 9529248 DOI: 10.1016/s0092-8674(00)81400-2] [Citation(s) in RCA: 1078] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The INK4a gene encodes two distinct growth inhibitors--the cyclin-dependent kinase inhibitor p16Ink4a, which is a component of the Rb pathway, and the tumor suppressor p19Arf, which has been functionally linked to p53. Here we show that p19Arf potently suppresses oncogenic transformation in primary cells and that this function is abrogated when p53 is neutralized by viral oncoproteins and dominant-negative mutants but not by the p53 antagonist MDM2. This finding, coupled with the observations that p19Arf and MDM2 physically interact and that p19Rrf blocks MDM2-induced p53 degradation and transactivational silencing, suggests that p19Arf functions mechanistically to prevent MDM2's neutralization of p53. Together, our findings ascribe INK4a's potent tumor suppressor activity to the cooperative actions of its two protein products and their relation to the two central growth control pathways, Rb and p53.
Collapse
Affiliation(s)
- J Pomerantz
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Lavinsky RM, Jepsen K, Heinzel T, Torchia J, Mullen TM, Schiff R, Del-Rio AL, Ricote M, Ngo S, Gemsch J, Hilsenbeck SG, Osborne CK, Glass CK, Rosenfeld MG, Rose DW. Diverse signaling pathways modulate nuclear receptor recruitment of N-CoR and SMRT complexes. Proc Natl Acad Sci U S A 1998; 95:2920-5. [PMID: 9501191 PMCID: PMC19670 DOI: 10.1073/pnas.95.6.2920] [Citation(s) in RCA: 452] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Several lines of evidence indicate that the nuclear receptor corepressor (N-CoR) complex imposes ligand dependence on transcriptional activation by the retinoic acid receptor and mediates the inhibitory effects of estrogen receptor antagonists, such as tamoxifen, suppressing a constitutive N-terminal, Creb-binding protein/coactivator complex-dependent activation domain. Functional interactions between specific receptors and N-CoR or SMRT corepressor complexes are regulated, positively or negatively, by diverse signal transduction pathways. Decreased levels of N-CoR correlate with the acquisition of tamoxifen resistance in a mouse model system for human breast cancer. Our data suggest that N-CoR- and SMRT-containing complexes act as rate-limiting components in the actions of specific nuclear receptors, and that their actions are regulated by multiple signal transduction pathways.
Collapse
Affiliation(s)
- R M Lavinsky
- Howard Hughes Medical Institute, Department and School of Medicine, University of California at San Diego, La Jolla, CA 92093-0648, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Emiliani S, Fischle W, Van Lint C, Al-Abed Y, Verdin E. Characterization of a human RPD3 ortholog, HDAC3. Proc Natl Acad Sci U S A 1998; 95:2795-800. [PMID: 9501169 PMCID: PMC19648 DOI: 10.1073/pnas.95.6.2795] [Citation(s) in RCA: 251] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/1997] [Accepted: 12/29/1997] [Indexed: 02/06/2023] Open
Abstract
Histone acetylation levels in cells result from a dynamic equilibrium between competing histone acetylases and deacetylases. Changes in histone acetylation levels occur during both transcriptional activation and silencing. Cloning of the cDNA for a human histone deacetylase (HDAC1) has shown that it represents a human ortholog of the yeast transcriptional regulator RPD3. We have screened the expressed sequence tag database (National Center for Biotechnology Information) with the yeast RPD3 sequence and identified a human ortholog of RPD3, HDAC3. This cDNA encodes a protein of 428 amino acids with 58% sequence identity with HDAC1p. By using a specific polyclonal antiserum recognizing the C-terminal domain of HDAC3p and Western blotting, we detected a single approximately 49-kDa band in several tumor cell lines. HDAC3p is expressed predominantly in the nuclear compartment. Immunoprecipitation experiments with either an antiserum against HDAC3p or an anti-FLAG antiserum and a flagged HDAC3 cDNA showed that HDAc3p exhibits deacetylase activity both on free histones and on purified nucleosomes. This deacetylase activity is inhibited by trichostatin, trapoxin, and butyrate in vitro to the same degree as the deacetylase activity associated to HDAC1p. These observations identify another member of a growing family of human HDAC genes.
Collapse
Affiliation(s)
- S Emiliani
- Picower Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | | | |
Collapse
|
230
|
Li Y, Kang J, Horwitz MS. Interaction of an adenovirus E3 14.7-kilodalton protein with a novel tumor necrosis factor alpha-inducible cellular protein containing leucine zipper domains. Mol Cell Biol 1998; 18:1601-10. [PMID: 9488477 PMCID: PMC108875 DOI: 10.1128/mcb.18.3.1601] [Citation(s) in RCA: 150] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/1997] [Accepted: 11/14/1997] [Indexed: 02/06/2023] Open
Abstract
Early region 3 (E3) of group C human adenoviruses (Ad) encodes several inhibitors of tumor necrosis factor alpha (TNF-alpha) cytolysis, including an E3 14.7-kDa protein (E3-14.7K) and a heterodimer containing two polypeptides of 10.4 and 14.5 kDa. To understand the mechanism by which the viral proteins inhibit TNF-alpha functions, the E3-14.7K protein was used to screen a HeLa cell cDNA library to search for interacting proteins in the yeast two-hybrid system. A novel protein containing multiple leucine zipper domains without any significant homology with any known protein was identified and has been named FIP-2 (for 14.7K-interacting protein). FIP-2 interacted with E3-14.7K both in vitro and in vivo. It colocalized with Ad E3-14.7K in the cytoplasm, especially near the nuclear membrane, and caused redistribution of the viral protein. FIP-2 by itself does not cause cell death; however, it can reverse the protective effect of E3-14.7K on cell killing induced by overexpression of the intracellular domain of the 55-kDa TNF receptor or by RIP, a death protein involved in the TNF-alpha and Fas apoptosis pathways. Deletion analysis indicates that the reversal effect of FIP-2 depends on its interaction with E3-14.7K. Three major mRNA forms of FIP-2 have been detected in multiple human tissues, and expression of the transcripts was induced by TNF-alpha treatment in a time-dependent manner in two different cell lines. FIP-2 has consensus sequences for several potential posttranslational modifications. These data suggest that FIP-2 is one of the cellular targets for Ad E3-14.7K and that its mechanism of affecting cell death involves the TNF receptor, RIP, or a downstream molecule affected by either of these two molecules.
Collapse
Affiliation(s)
- Y Li
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | |
Collapse
|
231
|
Berger A, Cultaro CM, Segal S, Spiegel S. The potent lipid mitogen sphingosylphosphocholine activates the DNA binding activity of upstream stimulating factor (USF), a basic helix-loop-helix-zipper protein. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1390:225-36. [PMID: 9507145 DOI: 10.1016/s0005-2760(97)00180-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We previously demonstrated that the sphingolipid, sphingosylphosphocholine (SPC) increased DNA binding activity of AP-1 proteins accompanying cellular proliferation. Herein, the effects of SPC on DNA binding activity and transcription of the basic, helix-loop-helix, leucine zipper (bHLH-ZIP) proteins Myc, Max, and USF were investigated because they regulate genes involved in mitogenesis. E-box (CACGTG) DNA binding proteins were detected by electrophoretic mobility shift assays in nuclear extracts from Swiss 3T3 fibroblasts. The slowest migrating complex (complex I) increased within 1-3 min after treatment with SPC, remained elevated for 10 min, and increased again after 12 h. Complexes I and II contained USF-1 and USF-2 proteins, and complex I migrated similarly to recombinant USF-1 protein/DNA complex. Treatment of nuclear extracts with alkaline phosphatase decreased these complexes suggesting USF might be a phosphoprotein, post-translationally modified by SPC. max and usf-1 mRNA levels were unaffected by SPC treatment. In contrast, c-myc mRNA was rapidly elevated, reached maximum levels at 0.5-1 h, and showed an additional increase after 12 h, just preceding S phase. Thus, certain bHLH-ZIP transcription factors may be involved in cell growth regulation by SPC.
Collapse
Affiliation(s)
- A Berger
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
232
|
Foley KP, McArthur GA, Quéva C, Hurlin PJ, Soriano P, Eisenman RN. Targeted disruption of the MYC antagonist MAD1 inhibits cell cycle exit during granulocyte differentiation. EMBO J 1998; 17:774-85. [PMID: 9451002 PMCID: PMC1170426 DOI: 10.1093/emboj/17.3.774] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The switch from transcriptionally activating MYC-MAX to transcriptionally repressing MAD1-MAX protein heterodimers has been correlated with the initiation of terminal differentiation in many cell types. To investigate the function of MAD1-MAX dimers during differentiation, we disrupted the Mad1 gene by homologous recombination in mice. Analysis of hematopoietic differentiation in homozygous mutant animals revealed that cell cycle exit of granulocytic precursors was inhibited following the colony-forming cell stage, resulting in increased proliferation and delayed terminal differentiation of low proliferative potential cluster-forming cells. Surprisingly, the numbers of terminally differentiated bone marrow and peripheral blood granulocytes were essentially unchanged in Mad1 null mice. This imbalance between the frequencies of precursor and mature granulocytes was correlated with a compensatory decrease in granulocytic cluster-forming cell survival under apoptosis-inducing conditions. In addition, recovery of the peripheral granulocyte compartment following bone marrow ablation was significantly enhanced in Mad1 knockout mice. Two Mad1-related genes, Mxi1 and Mad3, were found to be expressed ectopically in adult spleen, indicating that functional redundancy and cross-regulation between MAD family members may allow for apparently normal differentiation in the absence of MAD1. These findings demonstrate that MAD1 regulates cell cycle withdrawal during a late stage of granulocyte differentiation, and suggest that the relative levels of MYC versus MAD1 mediate a balance between cell proliferation and terminal differentiation.
Collapse
Affiliation(s)
- K P Foley
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North-Mailstop A2-025, P.O. Box 19024, Seattle, WA 98109-1024, USA
| | | | | | | | | | | |
Collapse
|
233
|
Abstract
PURPOSE This review summarizes the molecular genetics of childhood leukemias, with emphasis on pathogenesis and clinical applications. DESIGN We first describe the most common genetic events that occur in pediatric acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic myeloid leukemia (CML). We then illustrate how these molecular alterations may be used to alter therapy. RESULTS In childhood ALL, the TEL-AML1 fusion and hyperdiploidy are both associated with excellent treatment outcomes and therefore identify patients who may be candidates for less intensive therapy. In contrast, MLL gene rearrangements and the BCR-ABL fusion confer a poor prognosis; these patients may be best treated by allogeneic bone marrow transplantation in first remission. CONCLUSIONS Although clinical features are important prognostic indicators, genetic alterations of leukemic blasts may be better predictors of outcome for acute leukemia patients. We therefore favor risk-adapted therapy based on classification schemes that incorporate both genetic and clinical features.
Collapse
Affiliation(s)
- J E Rubnitz
- Department of Hematology/Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-2794, USA
| | | |
Collapse
|
234
|
Wang H, Van Den Bergh F, Spencer E, Wilcox K, Herman T. Use of a two-hybrid system to identify mutations in Max that confer increased affinity for Myc. DNA Cell Biol 1997; 16:1277-88. [PMID: 9407000 DOI: 10.1089/dna.1997.16.1277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A yeast two-hybrid system was used to identify mutants of Max that exhibit an increased affinity for Myc. Truncated forms of the Max helix-loop-helix/leucine zipper motif (HLH/Zip) were first expressed in a two- hybrid system in which the bait protein was the HLH/Zip motif of Myc. Deletion of amino acids both amino-terminal and carboxy-terminal to the leucine zipper of Max reduced Myc/Max heterodimer formation as evidenced by a 160-fold reduction in the expression of the lacZ gene. A library of partially randomized sequences encoding this minimal leucine zipper of Max was then screened using the two-hybrid system. Mutant forms of the Max leucine zipper were identified whose affinities for Myc, as measured by beta-galactosidase activity in yeast lysates, were from 8- to 200-fold greater than the wild-type Max zipper. These Max mutants were shown to interact specifically with Myc and not with wild-type Max. Of 29 mutants analyzed, all had a unique amino acid sequence. This result illustrates the value of a genetic screen in the identification of a collection of mutant forms of the Max leucine zipper whose structures would not have been predicted based on principles of structure-based design.
Collapse
Affiliation(s)
- H Wang
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | | | | | |
Collapse
|
235
|
Dang C, Lewis B. Role of Oncogenic Transcription Factor c-Myc in Cell Cycle Regulation, Apoptosis and Metabolism. J Biomed Sci 1997; 4:269-278. [PMID: 12386373 DOI: 10.1007/bf02258350] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The myc gene was initially discovered as a prototypical retrovirally transduced oncogene. Over the decades, abundant evidence has emerged to support a causal role for the activated cellular gene, c-myc, in animal and human tumors. The gene encodes an oncogenic helix-loop-helix leucine zipper transcription factor that acts as a heterodimer with its partner protein, Max, to activate genes regulating the cell cycle machinery as well as critical metabolic enzymes. The additional ability of c-Myc to repress transcription of differentiation-related genes suggest that c-Myc is a central and key molecular integrator of cell proliferation, differentiation and metabolism.
Collapse
Affiliation(s)
- C.V. Dang
- Departments of Medicine, Oncology, Pathology, and Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Md., USA
| | | |
Collapse
|
236
|
Peterson AJ, Kyba M, Bornemann D, Morgan K, Brock HW, Simon J. A domain shared by the Polycomb group proteins Scm and ph mediates heterotypic and homotypic interactions. Mol Cell Biol 1997; 17:6683-92. [PMID: 9343432 PMCID: PMC232522 DOI: 10.1128/mcb.17.11.6683] [Citation(s) in RCA: 117] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The Sex comb on midleg (Scm) and polyhomeotic (ph) proteins are members of the Polycomb group (PcG) of transcriptional repressors. PcG proteins maintain differential patterns of homeotic gene expression during development in Drosophila flies. The Scm and ph proteins share a homology domain with 38% identity over a length of 65 amino acids, termed the SPM domain, that is located at their respective C termini. Using the yeast two-hybrid system and in vitro protein-binding assays, we show that the SPM domain mediates direct interaction between Scm and ph. Binding studies with isolated SPM domains from Scm and ph show that the domain is sufficient for these protein interactions. These studies also show that the Scm-ph and Scm-Scm domain interactions are much stronger than the ph-ph domain interaction, indicating that the isolated domain has intrinsic binding specificity determinants. Analysis of site-directed point mutations identifies residues that are important for SPM domain function. These binding properties, predicted alpha-helical secondary structure, and conservation of hydrophobic residues prompt comparisons of the SPM domain to the helix-loop-helix and leucine zipper domains used for homotypic and heterotypic protein interactions in other transcriptional regulators. In addition to in vitro studies, we show colocalization of the Scm and ph proteins at polytene chromosome sites in vivo. We discuss the possible roles of the SPM domain in the assembly or function of molecular complexes of PcG proteins.
Collapse
Affiliation(s)
- A J Peterson
- Department of Biochemistry, University of Minnesota, St. Paul 55108, USA
| | | | | | | | | | | |
Collapse
|
237
|
Abstract
A variety of genes, including c-myc, are activated by chemical toxicants in vivo and in vitro. Although enforced c-myc expression induces apoptosis after withdrawing survival factors, it is not clear if activation of the endogenous c-myc gene is an apoptotic signal after toxicant exposure. The renal tubular epithelium is a target for many toxicants. c-myc expression is activated by tubular damage. In quiescent LLC-PK1 renal epithelial cells, c-myc but not max or mad mRNA is induced by the nephrotoxicant S-(1,2-dichlorovinyl)-L-cysteine (DCVC). The kinetics of DCVC-induced c-myc expression and apoptosis suggested an association between cell death and prolonged activation of c-myc expression after toxicant exposure. Accordingly, prolonged activation of an estrogen receptor-Myc fusion construct, but not a construct in which a c-Myc transactivation domain had been deleted, was sufficient to induce apoptosis in LLC-PK1 cells. Moreover, under conditions in which necrosis was the predominant cell death pathway caused by DCVC in parental cells, overexpressing c-myc biased the cell death pathway toward apoptosis. DCVC also induced ornithine decarboxylase (odc) mRNA and activated the odc promoter. Activation of the odc promoter by DCVC required consensus c-Myc-Max binding sites in odc intron 1. Inhibiting ODC activity with alpha-difluoromethylornithine delayed DCVC-induced cell death. Therefore, odc is a target gene in the DCVC apoptotic pathway involving c-myc activation and contributes to apoptosis. Finally, a structurally related cytotoxic but nongenotoxic analog of DCVC did not induce c-myc and did not activate the odc promoter or induce apoptosis. The data support the hypothesis that activation of apoptotic cell death in quiescent renal epithelial cells involves induction of c-myc. This is the first study to demonstrate that c-myc induction by a specific nephrotoxicant leads to gene activation and cell death.
Collapse
Affiliation(s)
- Y Zhan
- W. Alton Jones Cell Science Center, Lake Placid, New York 12946, USA
| | | | | |
Collapse
|
238
|
Cultraro CM, Bino T, Segal S. Regulated expression and function of the c-Myc antagonist, Mad1, during a molecular switch from proliferation to differentiation. Curr Top Microbiol Immunol 1997; 224:149-58. [PMID: 9308238 DOI: 10.1007/978-3-642-60801-8_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- C M Cultraro
- NCI-Navy Medical Oncology Branch, National Cancer Institute, NIH, Bethesds, MD, USA
| | | | | |
Collapse
|
239
|
Hurlin PJ, Qúeva C, Eisenman RN. Mnt: a novel Max-interacting protein and Myc antagonist. Curr Top Microbiol Immunol 1997; 224:115-21. [PMID: 9308234 DOI: 10.1007/978-3-642-60801-8_11] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have identified a novel Max-binding protein, Mnt, which belongs to neither the Myc nor the Mad families (Hurlin et al. 1997). Mnt interacts with Max in vivo and functions as a transcriptional repressor of reporter genes containing promoter-proximal CACGTG sites. Mnt:Max complexes also efficiently suppress Myc-dependent activation from the same promoter. Transcription repression by Mnt maps to a 13 amino acid N-terminal region related to the Sin3 interaction domain (SID) of Mad proteins. This region of Mnt mediates interaction with mSin3 corepressor proteins and its deletion converts Mnt from a repressor to an activator and from a suppressor of Myc-dependent transformation to a cooperating oncogene. This latter result suggests that Mnt and Myc regulate an overlapping set of target genes in vivo. Expression of mnt RNA is observed in many tissues and in both proliferating and differentiating cells. Likewise, Mnt protein is expressed in many proliferating cell types in culture where both Myc:Max and Mnt:Max complexes are detected. An exception is P19 embryonal carcinoma cells, where Mnt is expressed and in a complex with Max, but Myc proteins are not detected. Mnt is likely to be a key regulator of Myc activities in vivo and, in addition, may possess Myc-independent functions.
Collapse
Affiliation(s)
- P J Hurlin
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98104, USA
| | | | | |
Collapse
|
240
|
Schreiber-Agus N, Alland L, Muhle R, Goltz J, Chen K, Stevens L, Stein D, DePinho RA. A biochemical and biological analysis of Myc superfamily interactions. Curr Top Microbiol Immunol 1997; 224:159-68. [PMID: 9308239 DOI: 10.1007/978-3-642-60801-8_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- N Schreiber-Agus
- Dept. of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
241
|
Larsson LG, Bahram F, Wu S, Oberg F, Nilsson K, Lüscher B. Cytokine-induced inhibition of Myc activity in monocytic cells. Curr Top Microbiol Immunol 1997; 224:191-200. [PMID: 9308242 DOI: 10.1007/978-3-642-60801-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- L G Larsson
- Department of Pathology, University of Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
242
|
Potter M, Marcu KB. The c-myc story: where we've been, where we seem to be going. Curr Top Microbiol Immunol 1997; 224:1-17. [PMID: 9308224 DOI: 10.1007/978-3-642-60801-8_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- M Potter
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | |
Collapse
|
243
|
Hassig CA, Schreiber SL. Nuclear histone acetylases and deacetylases and transcriptional regulation: HATs off to HDACs. Curr Opin Chem Biol 1997; 1:300-8. [PMID: 9667866 DOI: 10.1016/s1367-5931(97)80066-x] [Citation(s) in RCA: 292] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Reversible acetylation of lysines on the amino-terminal tails of nucleosomal histones is correlated with changes in chromatin structure and transcription. The recent characterization of enzymes directly responsible for regulating histone acetylation and deacetylation and the cloning of their encoding cDNAs have provided insights into the possible functional and regulatory mechanisms of these classes of molecules. Nuclear histone acetylases have been shown to be transcriptional coactivators and coactivator-associated proteins, while histone deacetylases have been identified as components of nuclear co-repressor complexes. These findings confirm previous studies linking histone acetylation and transcriptional regulation.
Collapse
Affiliation(s)
- C A Hassig
- Howard Hughes Medical Institute, Harvard University Department of Chemistry, 12 Oxford Street, Cambridge, MA 02138, USA. hassig@slsiris harvard.edu
| | | |
Collapse
|
244
|
Ladomery M, Lyons S, Sommerville J. Xenopus HDm, a maternally expressed histone deacetylase, belongs to an ancient family of acetyl-metabolizing enzymes. Gene X 1997; 198:275-80. [PMID: 9370292 DOI: 10.1016/s0378-1119(97)00325-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Modification of core histones can alter chromatin structure, facilitating the activation and repression of genes. A key example is the acetylation of N-terminal lysines of the core histones. Recently, the mammalian histone deacetylase HD1 was cloned from Jurkat T cells, and shown to be 60% identical to the yeast global gene regulator Rpd3 (Taunton et al., 1996). Here we report the cloning of HDm, a maternally expressed putative deposition histone deacetylase from Xenopus laevis. Comparison of the amino acid sequences of histone deacetylases from diverse eukaryotes shows high levels of identity within a putative enzyme core region. Further alignment with other types of protein: acetoin-utilizing enzymes from eubacteria; acetylpolyamine hydrolases from mycoplasma and cyanobacteria; and a protein of unknown function from an archaebacterium, reveals an apparently conserved core, and suggests that histone deacetylases belong to an ancient family of enzymes with related functions.
Collapse
Affiliation(s)
- M Ladomery
- Division of Cell and Molecular Biology, School of Biological and Medical Sciences, University of St Andrews, Fife, Scotland, UK
| | | | | |
Collapse
|
245
|
Dhordain P, Albagli O, Lin RJ, Ansieau S, Quief S, Leutz A, Kerckaert JP, Evans RM, Leprince D. Corepressor SMRT binds the BTB/POZ repressing domain of the LAZ3/BCL6 oncoprotein. Proc Natl Acad Sci U S A 1997; 94:10762-7. [PMID: 9380707 PMCID: PMC23478 DOI: 10.1073/pnas.94.20.10762] [Citation(s) in RCA: 294] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The LAZ3/BCL6 (lymphoma-associated zinc finger 3/B cell lymphomas 6) gene frequently is altered in non-Hodgkin lymphomas. It encodes a sequence-specific DNA binding transcriptional repressor that contains a conserved N-terminal domain, termed BTB/POZ (bric-à-brac tramtrack broad complex/pox viruses and zinc fingers). Using a yeast two-hybrid screen, we show here that the LAZ3/BCL6 BTB/POZ domain interacts with the SMRT (silencing mediator of retinoid and thyroid receptor) protein. SMRT originally was identified as a corepressor of unliganded retinoic acid and thyroid receptors and forms a repressive complex with a mammalian homolog of the yeast transcriptional repressor SIN3 and the HDAC-1 histone deacetylase. Protein binding assays demonstrate that the LAZ3/BCL6 BTB/POZ domain directly interacts with SMRT in vitro. Furthermore, DNA-bound LAZ3/BCL6 recruits SMRT in vivo, and both overexpressed proteins completely colocalize in nuclear dots. Finally, overexpression of SMRT enhances the LAZ3/BCL6-mediated repression. These results define SMRT as a corepressor of LAZ3/BCL6 and suggest that LAZ3/BCL6 and nuclear hormone receptors repress transcription through shared mechanisms involving SMRT recruitment and histone deacetylation.
Collapse
Affiliation(s)
- P Dhordain
- U124 Institut National de la Santé et de la Recherche Médicale, Institut de Recherches sur le Cancer de Lille Place de Verdun, F-59045 Lille Cedex France.
| | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Abstract
'The amino termini of histones extend from the nucleosomal core and are modified by acetyltransferases and deacetylases during the cell cycle. These acetylation patterns may direct histone assembly and help regulate the unfolding and activity of genes.
Collapse
Affiliation(s)
- M Grunstein
- Department of Biological Chemistry, UCLA School of Medicine and the Molecular Biology Institute, University of California, Los Angeles 90095, USA
| |
Collapse
|
247
|
Bartl S, Taplick J, Lagger G, Khier H, Kuchler K, Seiser C. Identification of mouse histone deacetylase 1 as a growth factor-inducible gene. Mol Cell Biol 1997; 17:5033-43. [PMID: 9271381 PMCID: PMC232354 DOI: 10.1128/mcb.17.9.5033] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Reversible acetylation of core histones plays an important role in transcriptional regulation, cell cycle progression, and developmental events. The acetylation state of histones is controlled by the activities of acetylating and deacetylating enzymes. By using differential mRNA display, we have identified a mouse histone deacetylase gene, HD1, as an interleukin-2-inducible gene in murine T cells. Sequence alignments revealed that murine HD1 is highly homologous to the yeast RPD3 pleiotropic transcriptional regulator. Indirect immunofluorescence microscopy proved that mouse HD1 is a nuclear protein. When expressed in yeast, murine HD1 was also detected in the nucleus, although it failed to complement the rpd3delta deletion phenotype. HD1 mRNA expression was low in G0 mouse cells but increased when the cells crossed the G1/S boundary after growth stimulation. Immunoprecipitation experiments and functional in vitro assays showed that HD1 protein is associated with histone deacetylase activity. Both HD1 protein levels and total histone deacetylase activity increased upon interleukin-2 stimulation of resting B6.1 cells. When coexpressed with a luciferase reporter construct, HD1 acted as a negative regulator of the Rous sarcoma virus enhancer/promoter. HD1 overexpression in stably transfected Swiss 3T3 cells caused a severe delay during the G2/M phases of the cell cycle. Our results indicate that balanced histone acetylation/deacetylation is crucial for normal cell cycle progression of mammalian cells.
Collapse
Affiliation(s)
- S Bartl
- Institute of Molecular Biology, University of Vienna, Vienna Biocenter, Austria
| | | | | | | | | | | |
Collapse
|
248
|
Abstract
Mad proteins are transcriptional repressors that antagonize transcriptional activation and transformation by Myc oncoprotein; recent findings suggest that they repress transcription by recruiting histone deacetylases to target sites on DNA.
Collapse
Affiliation(s)
- A Kiermaier
- Zentrum für Molekulare Biologie, Heidelberg, Germany
| | | |
Collapse
|
249
|
Zhang H, Fan S, Prochownik EV. Distinct roles for MAX protein isoforms in proliferation and apoptosis. J Biol Chem 1997; 272:17416-24. [PMID: 9211884 DOI: 10.1074/jbc.272.28.17416] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
MAX is a basic helix-loop-helix-leucine zipper protein that plays a central role in the transcriptional control of Myc oncoproteins. MYC-MAX heterodimers stimulate transcription, whereas MAX homodimers, or heterodimers between MAX and members of the MAD family of basic helix-loop-helix-leucine zipper proteins, repress transcription. Max exists in two major isomeric forms, MAX(L) and MAX(S), which differ from one another only by a 9-amino acid insertion/deletion. We show here that MAX(L) is much more effective at homodimeric DNA binding than MAX(S). In NIH3T3 cells, MAX(L) was able to repress a c-Myc-responsive reporter gene whereas MAX(S) either stimulated the reporter gene or had little effect on its expression. In comparison to control cell lines or those stably over-expressing MAX(S), MAX(L)-over-expressing cell lines showed reduced expression of transiently expressed or endogenous c-Myc responsive genes, grew more slowly, possessed a higher growth factor requirement, and showed accelerated apoptosis following growth factor deprivation. Differential effects on growth and apoptosis represent two previously unrecognized properties of MAX proteins. These can at least partly be explained by the differences in their DNA binding abilities and their effects on target gene expression.
Collapse
Affiliation(s)
- H Zhang
- Department of Molecular Genetics and Biochemistry, The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
250
|
Pusch O, Soucek T, Hengstschläger-Ottnad E, Bernaschek G, Hengstschläger M. Cellular targets for activation by c-Myc include the DNA metabolism enzyme thymidine kinase. DNA Cell Biol 1997; 16:737-47. [PMID: 9212167 DOI: 10.1089/dna.1997.16.737] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Although a remarkable number of genes has been identified that are either activated or repressed via c-Myc, only few of them obviously contribute to Myc's biological effect--the induction of proliferation. We found that in logarithmically growing cells overexpression of Myc specifically induces thymidine kinase (TK) mRNA expression and enzyme activity, whereas loss of one allele of Myc causes downregulation of this enzyme. We show that activation of Myc triggers high levels of this normally strictly S-phase-regulated DNA metabolism enzyme in serum arrested G0 cells and causes high and constant levels of TK expression throughout the entire ongoing cell cycle. Induction of TK by Myc requires an intact transcriptional activation domain. Myc-induced deregulation of this enzyme is paralleled by alterations of protein binding at the E2F-site of the TK promoter. We further show that cell growth arrest by the cyclin-dependent kinase inhibitor p16 is abrogated by overexpression of Myc and that co-overexpression of p16 cannot inhibit the Myc-induced up-regulation of TK expression. Our data demonstrate TK to be a cellular target of Myc independently of the status of cell proliferation and provide evidence that the transcription factor E2F might be involved in this process.
Collapse
Affiliation(s)
- O Pusch
- Obstetrics and Gynecology, University of Vienna, Department of Prenatal Diagnosis and Therapy, Austria
| | | | | | | | | |
Collapse
|