Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795443

ABSTRACT

The increase in AP-1 activity is a hallmark of cell transformation by tyrosine kinases. Previously, we reported that blocking AP-1 using the c-Jun dominant negative mutant TAM67 induced senescence, adipogenesis, or apoptosis in v-Src-transformed chicken embryo fibroblasts (CEFs) whereas inhibition of JunD by short hairpin RNA (shRNA) specifically induced apoptosis. To investigate the role of AP-1 in Src-mediated transformation, we undertook a gene profiling study to characterize the transcriptomes of v-Src-transformed CEFs expressing either TAM67 or the JunD shRNA. Our study revealed a cluster of 18 probe sets upregulated exclusively in response to AP-1/JunD impairment and v-Src transformation. Four of these probe sets correspond to genes involved in the interferon pathway. One gene in particular, death-associated protein kinase 1 (DAPK1), is a C/EBPß-regulated mediator of apoptosis in gamma interferon (IFN-γ)-induced cell death. Here, we show that inhibition of DAPK1 abrogates cell death in v-Src-transformed cells expressing the JunD shRNA. Chromatin immunoprecipitation data indicated that C/EBPß was recruited to the DAPK1 promoter while the expression of a dominant negative mutant of C/EBPß abrogated the induction of DAPK1 in response to the inhibition of AP-1. In contrast, as determined by chromatin immunoprecipitation (ChIP) assays, JunD was not detected on the DAPK1 promoter under any conditions, suggesting that JunD promotes survival by indirectly antagonizing the expression of DAPK1 in v-Src transformed cells. IMPORTANCE: Transformation by the v-Src oncoprotein causes extensive changes in gene expression in primary cells such as chicken embryo fibroblasts. These changes, determining the properties of transformed cells, are controlled in part at the transcriptional level. Much attention has been devoted to transcription factors such as AP-1 and NF-κB and the control of genes associated with a more aggressive phenotype. In this report, we describe a novel mechanism of action determined by the JunD component of AP-1, a factor enhancing cell survival in v-Src-transformed cells. We show that the loss of JunD results in the aberrant activation of a genetic program leading to cell death. This program requires the activation of the tumor suppressor death-associated protein kinase 1 (DAPK1). Since DAPK1 is phosphorylated and inhibited by v-Src, these results highlight the importance of this kinase and the multiple mechanisms controlled by v-Src to antagonize the tumor suppressor function of DAPK1.


Subject(s)
Death-Associated Protein Kinases/genetics , Fibroblasts/metabolism , Oncogene Protein pp60(v-src)/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/genetics , Animals , Apoptosis/genetics , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Survival , Cells, Cultured , Chick Embryo , Chickens , Chromatin Immunoprecipitation , Death-Associated Protein Kinases/metabolism , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation , Oncogene Protein pp60(v-src)/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Transfection
2.
J Virol ; 85(13): 6725-35, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21507983

ABSTRACT

The activation of AP-1 is a hallmark of cell transformation by tyrosine kinases. In this study, we characterize the role of AP-1 proteins in the transformation of chicken embryo fibroblasts (CEF) by v-Src. In normal CEF, the expression of a dominant negative mutant of c-Jun (TAM67) induced senescence. In contrast, three distinct phenotypes were observed when TAM67 was expressed in v-Src-transformed CEF. While senescent cells were also present, the inhibition of AP-1 caused apoptosis in a fraction of the v-Src-transformed cells. In addition, cells containing lipid-rich vesicles accumulated, suggesting that a subpopulation of the v-Src-transformed cells underwent differentiation in response to the inhibition of AP-1. JunD and Fra-2 were the main components of this factor, while c-Jun accounted for a minor fraction of AP-1 in v-Src-transformed CEF. The downregulation of c-Jun expression by short hairpin RNA (shRNA) induced senescence in normal and v-Src-transformed cells. In contrast, a high incidence of apoptosis was caused by the downregulation of JunD, suggesting that it is required for the survival of v-Src-transformed CEF. Levels of the p53 tumor suppressor were elevated under conditions of JunD inhibition. Repression of p53 by shRNA enhanced the survival and anchorage-independent proliferation of v-Src-transformed CEF with JunD/AP-1 inhibition. The inhibition of Fra-2 had no visible phenotype in normal CEF but caused the appearance of lipid-rich vesicles in v-Src-transformed CEF. Therefore, AP-1 facilitated transformation by acting as a survival factor, by inhibiting premature entry into senescence, and by blocking the differentiation of v-Src-transformed CEF.


Subject(s)
Cell Transformation, Viral , Fibroblasts/virology , Gene Expression Regulation , Genes, src , Genetic Pleiotropy/physiology , Rous sarcoma virus/physiology , Transcription Factor AP-1/metabolism , Animals , Cell Line, Transformed , Chick Embryo , Enzyme Activation , Fibroblasts/metabolism , Fibroblasts/physiology , Fos-Related Antigen-2 , Genetic Pleiotropy/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/genetics
3.
PLoS One ; 5(11): e14049, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21124979

ABSTRACT

BACKGROUND: The multiple endocrine neoplasia type I gene functions as a tumor suppressor gene in humans and mouse models. In Drosophila melanogaster, mutants of the menin gene (Mnn1) are hypersensitive to mutagens or gamma irradiation and have profound defects in the response to several stresses including heat shock, hypoxia, hyperosmolarity and oxidative stress. However, it is not known if the function of menin in the stress response contributes to genome stability. The objective of this study was to examine the role of menin in the control of the stress response and genome stability. METHODOLOGY/PRINCIPAL FINDINGS: Using a test of loss-of-heterozygosity, we show that Drosophila strains lacking a functional Mnn1 gene or expressing a Mnn1 dsRNA display increased genome instability in response to non-lethal heat shock or hypoxia treatments. This is also true for strains lacking all Hsp70 genes, implying that a precise control of the stress response is required for genome stability. While menin is required for Hsp70 expression, the results of epistatic studies indicate that the increase in genome instability observed in Mnn1 lack-of-function mutants cannot be accounted for by mis-expression of Hsp70. Therefore, menin may promote genome stability by controlling the expression of other stress-responsive genes. In agreement with this notion, gene profiling reveals that Mnn1 is required for sustained expression of all heat shock protein genes but is dispensable for early induction of the heat shock response. CONCLUSIONS/SIGNIFICANCE: Mutants of the Mnn1 gene are hypersensitive to several stresses and display increased genome instability when subjected to conditions, such as heat shock, generally regarded as non-genotoxic. In this report, we describe a role for menin as a global regulator of heat shock gene expression and critical factor in the maintenance of genome integrity. Therefore, menin links the stress response to the control of genome stability in Drosophila melanogaster.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genomic Instability , Heat-Shock Response/genetics , Animals , Cluster Analysis , Drosophila melanogaster/metabolism , Gene Deletion , Gene Expression Profiling , Genome, Insect/genetics , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Hypoxia , Loss of Heterozygosity , Mice , Mutation , Oligonucleotide Array Sequence Analysis , RNA Interference
4.
BMC Cancer ; 10: 41, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20152043

ABSTRACT

BACKGROUND: Cell transformation by the Src tyrosine kinase is characterized by extensive changes in gene expression. In this study, we took advantage of several strains of the Rous sarcoma virus (RSV) to characterize the patterns of v-Src-dependent gene expression in two different primary cell types, namely chicken embryo fibroblasts (CEF) and chicken neuroretinal (CNR) cells. We identified a common set of v-Src regulated genes and assessed if their expression is associated with disease-free survival using several independent human tumor data sets. METHODS: CEF and CNR cells were infected with transforming, non-transforming, and temperature sensitive mutants of RSV to identify the patterns of gene expression in response to v-Src-transformation. Microarray analysis was used to measure changes in gene expression and to define a common set of v-Src regulated genes (CSR genes) in CEF and CNR cells. A clustering enrichment regime using the CSR genes and two independent breast tumor data-sets was used to identify a 42-gene aggressive tumor gene signature. The aggressive gene signature was tested for its prognostic value by conducting survival analyses on six additional tumor data sets. RESULTS: The analysis of CEF and CNR cells revealed that cell transformation by v-Src alters the expression of 6% of the protein coding genes of the genome. A common set of 175 v-Src regulated genes (CSR genes) was regulated in both CEF and CNR cells. Within the CSR gene set, a group of 42 v-Src inducible genes was associated with reduced disease- and metastasis-free survival in several independent patient cohorts with breast or lung cancer. Gene classes represented within this group include DNA replication, cell cycle, the DNA damage and stress responses, and blood vessel morphogenesis. CONCLUSION: By studying the v-Src-dependent changes in gene expression in two types of primary cells, we identified a set of 42 inducible genes associated with poor prognosis in breast and lung cancer. The identification of these genes provides a set of biomarkers of aggressive tumor behavior and a framework for the study of cancer cells characterized by elevated Src kinase activity.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, src , Oncogene Protein pp60(v-src)/biosynthesis , Animals , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Chick Embryo , Cluster Analysis , Cohort Studies , Disease-Free Survival , Fibroblasts/cytology , Humans , Lung Neoplasms/metabolism , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Prognosis , Retina/cytology
5.
Adv Exp Med Biol ; 668: 27-36, 2009.
Article in English | MEDLINE | ID: mdl-20175450

ABSTRACT

The cloning of the MEN1 gene in 1997 led to the characterization of menin, the protein behind the multiple endocrine neoplasia Type 1 syndrome. Menin, a novel nuclear protein with no homology to other gene products, is expressed ubiquitously. MEN1 missense mutations are dispersed along the coding region of the gene but are more common in the most conserved regions. Likewise, domains of protein interaction often correspond to the more conserved segments of menin. These protein interactions are generally facilitated by multiple domains or encompass a large portion of menin. The exception to this rule is a small stretch of amino acids mediating the interaction of menin with the mSin3A corepressor and histone deacetylase complexes. The C-terminal region of menin harbors several nuclear localization signals that play redundant functions in the localization of menin to the nuclear compartment. The nuclear localization signals are also important for the interaction of menin with the nuclear matrix. Menin is the target of several kinases and a candidate substrate of the ATM/ATR kinases, implying a role for this tumor suppressor in the DNA damage response. Menin is highly conserved from Drosophila to human but is absent in the nematode and in yeast.


Subject(s)
Multiple Endocrine Neoplasia Type 1 , Proto-Oncogene Proteins , Amino Acid Sequence , Animals , Exons , Humans , Introns , Molecular Sequence Data , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/physiopathology , Mutation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
6.
Neurochem Int ; 49(5): 494-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16782232

ABSTRACT

The p38 members of the mitogen-activated protein kinase (MAPK) superfamily are activated by both environmental stress and endogenous signals, and may have either permissive or inhibitory roles upon both cell proliferation and cell death in the retina. We have previously shown that anisomycin, a protein synthesis inhibitor, and 2-aminopurine, a specific inhibitor of the double stranded-RNA dependent protein kinase, block apoptosis of ganglion cells induced by axotomy, and induce apoptosis of cells in the neuroblastic layer in developing rat retina. Using a specific inhibitor, we found that p38-stress activated MAP kinase is required for the death of post-mitotic cells induced by anisomycin, but not for the death of proliferating cells induced by 2-aminopurine, nor of axon-damaged retinal ganglion cells. We also show that p38 activation occurs either upstream of or parallel to the requirement for cyclic AMP to block apoptosis of post-mitotic cells, since the cyclic AMP-producing agent forskolin did not prevent p38 phosphorylation induced by anisomycin. Finally, the lack of immunostaining for phospho-p38 in apoptotic profiles suggests that p38 activation does not kill retinal cells directly, but more likely through the mediation of neighboring cells.


Subject(s)
Apoptosis/physiology , Retina/cytology , p38 Mitogen-Activated Protein Kinases/physiology , 2-Aminopurine/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation , Enzyme Activation , Immunohistochemistry , Phosphorylation , Rats , Rats, Long-Evans , Retina/enzymology , Retina/growth & development , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Mol Cell Biol ; 25(22): 9960-72, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16260610

ABSTRACT

Menin, the product of the multiple endocrine neoplasia type I gene, has been implicated in several biological processes, including the control of gene expression and apoptosis, the modulation of mitogen-activated protein kinase pathways, and DNA damage sensing or repair. In this study, we have investigated the function of menin in the model organism Drosophila melanogaster. We show that Drosophila lines overexpressing menin or an RNA interference for this gene develop normally but are impaired in their response to several stresses, including heat shock, hypoxia, hyperosmolarity and oxidative stress. In the embryo subjected to heat shock, this impairment was characterized by a high degree of developmental arrest and lethality. The overexpression of menin enhanced the expression of HSP70 in embryos and interfered with its down-regulation during recovery at the normal temperature. In contrast, the inhibition of menin with RNA interference reduced the induction of HSP70 and blocked the activation of HSP23 upon heat shock, Menin was recruited to the Hsp70 promoter upon heat shock and menin overexpression stimulated the activity of this promoter in embryos. A 70-kDa inducible form of menin was expressed in response to heat shock, indicating that menin is also regulated in conditions of stress. The induction of HSP70 and HSP23 was markedly reduced or absent in mutant embryos harboring a deletion of the menin gene. These embryos, which did not express the heat shock-inducible form of menin, were also hypersensitive to various conditions of stress. These results suggest a novel role for menin in the control of the stress response and in processes associated with the maintenance of protein integrity.


Subject(s)
Drosophila Proteins/biosynthesis , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Acridine Orange/pharmacology , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , DNA/chemistry , DNA Damage , DNA Repair , Down-Regulation , Drosophila melanogaster , Fluorescent Dyes/pharmacology , Genotype , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Hot Temperature , Hypoxia , Immunohistochemistry , Mitosis , Mutation , Oxidative Stress , RNA Interference , Temperature , Time Factors , Trachea/metabolism , Trachea/pathology , Transgenes , beta-Galactosidase/metabolism
8.
J Biol Chem ; 278(44): 43846-54, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12896981

ABSTRACT

Chicken embryo fibroblasts (CEF) express several growth arrest-specific (GAS) gene products in G0. In contact-inhibited cells, the expression of the most abundant of these proteins, the p20K lipocalin, is activated at the transcriptional level by C/EBPbeta. In this report, we describe the role of C/EBPbeta in CEF proliferation. We show that the expression of a dominant negative mutant of C/EBPbeta (designated Delta184-C/EBPbeta) completely inhibited p20K expression at confluence and stimulated the proliferation of CEF without inducing transformation. Mouse embryo fibroblasts nullizygous for C/EBPbeta had a proliferative advantage over cells with one or two functional copies of this gene. C/EBP inhibition enhanced the expression of the three major components of AP-1 in cycling CEF, namely c-Jun, JunD, and Fra-2, and stimulated AP-1 activity. In contrast, the over-expression of C/EBPbeta caused a dramatic reduction in the levels of AP-1 proteins. Therefore, C/EBPbeta is a negative regulator of AP-1 expression and activity in CEF. The expression of cyclin D1 and cell proliferation were stimulated by the dominant negative mutant of C/EBPbeta but not in the presence of TAM67, a dominant negative mutant of c-Jun and AP-1. CEF over-expressing c-Jun, and to a lesser extent JunD and Fra-2, did not growth arrest at high cell density and did not express p20K. Therefore, AP-1 interfered with the action of C/EBPbeta at high cell density, indicating that these factors play opposing roles in the control of GAS gene expression and CEF proliferation.


Subject(s)
Blood Proteins/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Fibroblasts/metabolism , Gene Expression Regulation , Transcription Factor AP-1/physiology , Animals , Avian Proteins , Blood Proteins/metabolism , Blotting, Western , Cell Division , Cells, Cultured , Chick Embryo , Cyclin D1/metabolism , DNA Fragmentation , Genes, Dominant , Lipocalins , Mutation , Time Factors , Transcription Factor AP-1/metabolism
9.
J Neurobiol ; 56(2): 171-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12838582

ABSTRACT

The phosphoinositide-3-kinase (PI3K)/protein kinase B (PKB)/Bad signal transduction pathway is engaged in the control of apoptosis in many different cell types, particularly through phosphorylation of the Bcl-2 family protein Bad. We examined the involvement of this pathway in the control of programmed cell death in the retina of developing rats. PKB is constitutively phosphorylated in retinal tissue in vitro, whereas Bad was dephosphorylated both in Ser112 and Ser136. Cell death induced by either the PI3K inhibitor LY294002, or the general kinase inhibitor 2-aminopurine, were followed by PKB dephosphorylation, but PKB was not modulated during cell death induced by the protein synthesis inhibitor anisomycin. Treatment of retinal tissue cultures with forskolin, which increases intracellular levels of cAMP, partially blocked apoptosis induced by both anisomycin and 2-aminopurine, but not by LY294002, whereas forskolin invariably induced phosphorylation of Bad on both Ser112 and Ser136. The data suggest that Bad may be engaged in survival pathways in the immature retina, but pathways other than PI3K/PKB/Bad, and phosphorylation sites other than Ser112 and Ser136 in the Bad protein control cell survival in retinal tissue.


Subject(s)
Carrier Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Retina/cytology , Retina/physiology , Animals , Animals, Newborn , Cell Count , Cell Death/physiology , Cytoprotection/physiology , Immunoblotting , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Rats, Long-Evans , Serine/metabolism , Signal Transduction/physiology , bcl-Associated Death Protein
10.
J Biol Chem ; 277(19): 16426-32, 2002 May 10.
Article in English | MEDLINE | ID: mdl-11877423

ABSTRACT

The role of activating protein-1 (AP-1) in muscle cells is currently equivocal. While some studies propose that AP-1 is inhibitory for myogenesis, others implicate a positive role in this process. We tested whether this variation may be due to different properties of the AP-1 subunit composition in differentiating cells. Using Western analysis we show that c-Jun, Fra-2, and JunD are expressed throughout the time course of differentiation. Phosphatase assays indicate that JunD and Fra-2 are phosphorylated in muscle cells and that at least two isoforms of each are expressed in muscle cells. Electrophoretic mobility shift assays combined with antibody supershifts indicate the appearance of Fra-2 as a major component of the AP-1 DNA binding complex in differentiating cells. In this context it appears that Fra-2 heterodimerizes with c-Jun and JunD. Studying the c-jun enhancer in reporter gene assays we observed that the muscle transcription factors MEF2A and MyoD can contribute to robust transcriptional activation of the c-jun enhancer. In differentiating muscle cells mutation of the MEF2 site reduces transactivation of the c-jun enhancer and MEF2A is the predominant MEF2 isoform binding to this cis element. Transcriptional activation of an AP-1 site containing reporter gene (TRE-Luc) is enhanced under differentiation conditions compared with growth conditions in C2C12 muscle cells. Further studies indicate that Fra-2 containing AP-1 complexes can transactivate the MyoD enhancer/promoter. Thus, an AP-1 complex containing Fra-2 and c-Jun or JunD is consistent with muscle differentiation, indicating that AP-1 function during myogenesis is dependent on its subunit composition.


Subject(s)
Muscles/cytology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/physiology , Animals , Blotting, Western , COS Cells , Cell Differentiation , Cell Division , Cells, Cultured , DNA-Binding Proteins/metabolism , Fos-Related Antigen-2 , Genes, Reporter , HeLa Cells , Humans , Immunoblotting , Luciferases/metabolism , Models, Biological , Muscles/metabolism , Phosphorylation , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Proto-Oncogene Proteins c-jun/metabolism , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...