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










Publication year range
1.
Blood Adv ; 4(22): 5616-5630, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33186461

ABSTRACT

Multiple myeloma (MM) is a hematologic malignancy produced by a clonal expansion of plasma cells and characterized by abnormal production and secretion of monoclonal antibodies. This pathology exhibits an enormous heterogeneity resulting not only from genetic alterations but also from several epigenetic dysregulations. Here we provide evidence that Che-1/AATF (Che-1), an interactor of RNA polymerase II, promotes MM proliferation by affecting chromatin structure and sustaining global gene expression. We found that Che-1 depletion leads to a reduction of "active chromatin" by inducing a global decrease of histone acetylation. In this context, Che-1 directly interacts with histones and displaces histone deacetylase class I members from them. Strikingly, transgenic mice expressing human Che-1 in plasma cells develop MM with clinical features resembling those observed in the human disease. Finally, Che-1 downregulation decreases BRD4 chromatin accumulation to further sensitize MM cells to bromodomain and external domain inhibitors. These findings identify Che-1 as a promising target for MM therapy, alone or in combination with bromodomain and external domain inhibitors.


Subject(s)
Multiple Myeloma , Nuclear Proteins , Cell Proliferation , Chromatin , Humans , Multiple Myeloma/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics
2.
J Exp Clin Cancer Res ; 36(1): 32, 2017 02 18.
Article in English | MEDLINE | ID: mdl-28214471

ABSTRACT

BACKGROUND: Solid tumours are less oxygenated than normal tissues. Consequently, cancer cells acquire to be adapted to a hypoxic environment. The poor oxygenation of solid tumours is also a major indicator of an adverse cancer prognosis and leads to resistance to conventional anticancer treatments. We previously showed the involvement of Che-1/AATF (Che-1) in cancer cell survival under stress conditions. Herein we hypothesized that Che-1 plays a role in the response of cancer cells to hypoxia. METHODS: The human colon adenocarcinoma HCT116 and HT29 cell lines undepleted or depleted for Che-1 expression by siRNA, were treated under normoxic and hypoxic conditions to perform studies regarding the role of this protein in metabolic adaptation and cell proliferation. Che-1 expression was detected using western blot assays; cell metabolism was assessed by NMR spectroscopy and functional assays. Additional molecular studies were performed by RNA seq, qRT-PCR and ChIP analyses. RESULTS: Here we report that Che-1 expression is required for the adaptation of cells to hypoxia, playing an important role in metabolic modulation. Indeed, Che-1 depletion impacted on HIF-1α stabilization, thus downregulating the expression of several genes involved in the response to hypoxia and affecting glucose metabolism. CONCLUSIONS: We show that Che-1 a novel player in the regulation of HIF-1α in response to hypoxia. Notably, we found that Che-1 is required for SIAH-2 expression, a member of E3 ubiquitin ligase family that is involved in the degradation of the hydroxylase PHD3, the master regulator of HIF-1α stability.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Colorectal Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Repressor Proteins/genetics , Cell Hypoxia , Cell Proliferation , Colorectal Neoplasms/chemistry , Gene Expression Regulation, Neoplastic , Glucose/metabolism , HCT116 Cells , HT29 Cells , Humans , Protein Stability , Sequence Analysis, RNA
3.
Oncotarget ; 7(43): 70546-70558, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27655709

ABSTRACT

Multiple myeloma (MM) is a malignant disorder of plasma cells characterized by active production and secretion of monoclonal immunoglobulins (IgG), thus rendering cells prone to endoplasmic reticulum (ER) stress. For this reason, MM cell survival requires to maintain ER homeostasis at basal levels. Deptor is an mTOR binding protein, belonging to the mTORC1 and mTORC2 complexes. It was reported that Deptor is overexpressed in MM cells where it inhibits mTOR kinase activity and promotes cell survival by activating Akt signaling. Here we identify Deptor as a nuclear protein, able to bind DNA and regulate transcription in MM cells. In particular, we found that Deptor plays an important role in the maintenance of the ER network, sustaining the expression of several genes involved in this pathway. In agreement with this, Deptor depletion induces ER stress and synergizes the effect of the proteasome inhibitor bortezomib (Bz) in MM cells. These findings provide important new insights in the ER stress control in MM cells.


Subject(s)
Endoplasmic Reticulum , Homeostasis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Transgenic , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , RNA Interference
4.
EMBO J ; 34(9): 1214-30, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25770584

ABSTRACT

Mammalian target of rapamycin (mTOR) is a key protein kinase that regulates cell growth, metabolism, and autophagy to maintain cellular homeostasis. Its activity is inhibited by adverse conditions, including nutrient limitation, hypoxia, and DNA damage. In this study, we demonstrate that Che-1, a RNA polymerase II-binding protein activated by the DNA damage response, inhibits mTOR activity in response to stress conditions. We found that, under stress, Che-1 induces the expression of two important mTOR inhibitors, Redd1 and Deptor, and that this activity is required for sustaining stress-induced autophagy. Strikingly, Che-1 expression correlates with the progression of multiple myeloma and is required for cell growth and survival, a malignancy characterized by high autophagy response.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Multiple Myeloma/pathology , Repressor Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Survival , Female , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice, Nude , Multiple Myeloma/metabolism , Multiprotein Complexes/metabolism , Phosphorylation , Repressor Proteins/genetics , Stress, Physiological , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
J Biol Chem ; 288(32): 23348-57, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23798705

ABSTRACT

To combat threats posed by DNA damage, cells have evolved mechanisms, collectively termed DNA damage response (DDR). These mechanisms detect DNA lesions, signal their presence, and promote their repair. Centrosomes integrate G2/M checkpoint control and repair signals in response to genotoxic stress, acting as an efficient control mechanism when G2/M checkpoint function fails and mitosis begins in the presence of damaged DNA. Che-1 is an RNA polymerase II-binding protein involved in the regulation of gene transcription, induction of cell proliferation, and DDR. Here we provide evidence that in addition to its nuclear localization, Che-1 localizes at interphase centrosomes, where it accumulates following DNA damage or spindle poisons. We show that Che-1 depletion generates supernumerary centrosomes, multinucleated cells, and multipolar spindle formation. Notably, Che-1 depletion abolishes the ability of Chk1 to bind pericentrin and to localize at centrosomes, which, in its turn, deregulates the activation of centrosomal cyclin B-Cdk1 and advances entry into mitosis. Our results reinforce the notion that Che-1 plays an important role in DDR and that its contribution seems to be relevant for the spindle assembly checkpoint.


Subject(s)
Antigens/metabolism , Apoptosis Regulatory Proteins/metabolism , Centrosome/metabolism , Chromosomes, Human/metabolism , DNA Damage , Mitosis/physiology , Protein Kinases/metabolism , Repressor Proteins/metabolism , Antigens/genetics , Apoptosis Regulatory Proteins/genetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , Chromosomes, Human/genetics , Cyclin B/genetics , Cyclin B/metabolism , G2 Phase Cell Cycle Checkpoints/physiology , Humans , M Phase Cell Cycle Checkpoints/physiology , Protein Kinases/genetics , Repressor Proteins/genetics
6.
Cancer Cell ; 18(2): 122-34, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20708154

ABSTRACT

Che-1 is a RNA polymerase II binding protein involved in the regulation of gene transcription and, in response to DNA damage, promotes p53 transcription. In this study, we investigated whether Che-1 regulates mutant p53 expression. We found that Che-1 is required for sustaining mutant p53 expression in several cancer cell lines, and that Che-1 depletion by siRNA induces apoptosis both in vitro and in vivo. Notably, loss of Che-1 activates DNA damage checkpoint response and induces transactivation of p73. Therefore, these findings underline the important role that Che-1 has in survival of cells expressing mutant p53.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Cell Survival/physiology , DNA Damage , Repressor Proteins/physiology , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Repair/physiology , DNA-Binding Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics , RNA, Small Interfering , Repressor Proteins/genetics , Transplantation, Heterologous , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
7.
Hum Mol Genet ; 19(5): 752-60, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19965907

ABSTRACT

The absence of the cytoskeletal protein dystrophin results in Duchenne muscular dystrophy (DMD). The utrophin protein is the best candidate for dystrophin replacement in DMD patients. To obtain therapeutic levels of utrophin expression in dystrophic muscle, we developed an alternative strategy based on the use of artificial zinc finger transcription factors (ZF ATFs). The ZF ATF 'Jazz' was recently engineered and tested in vivo by generating a transgenic mouse specifically expressing Jazz at the muscular level. To validate the ZF ATF technology for DMD treatment we generated a second mouse model by crossing Jazz-transgenic mice with dystrophin-deficient mdx mice. Here, we show that the artificial Jazz protein restores sarcolemmal integrity and prevents the development of the dystrophic disease in mdx mice. This exclusive animal model establishes the notion that utrophin-based therapy for DMD can be efficiently developed using ZF ATF technology and candidates Jazz as a novel therapeutic molecule for DMD therapy.


Subject(s)
Muscular Dystrophy, Animal/therapy , Transcription Factors/genetics , Utrophin/genetics , Animals , Dystrophin/genetics , Dystrophin/metabolism , Female , Male , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Utrophin/metabolism , Zinc Fingers
8.
BMC Cell Biol ; 9: 56, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18840275

ABSTRACT

BACKGROUND: Chimeric proteins obtained by the fusion of a G protein-coupled receptor (GPCR) sequence to the N-terminus of the G protein alpha-subunit have been extensively used to investigate several aspects of GPCR signalling. Although both the receptor and the G protein generally maintain a fully functional state in such polypeptides, original observations made using a chimera between the beta2-adrenergic receptor (beta2AR) and Galphas indicated that the fusion to the alpha-subunit resulted in a marked reduction of receptor desensitization and down-regulation. To further investigate this phenomenon, we have compared the rates of internalization and recycling between wild-type and Galphas-fused beta2AR. RESULTS: The rate of agonist-induced internalization, measured as the disappearance of cell surface immunofluorescence in HEK293 cells permanently expressing N-terminus tagged receptors, was reduced three-fold by receptor-G protein fusion. However, both fused and non-fused receptors translocated to the same endocytic compartment, as determined by dual-label confocal analysis of cells co-expressing both proteins and transferrin co-localization. Receptor recycling, determined as the reversion of surface immunofluorescence following the addition of antagonist to cells that were previously exposed to agonist, markedly differed between wild-type and fused receptors. While most of the internalized beta2AR returned rapidly to the plasma membrane, beta2AR-Galphas did not recycle, and the observed slow recovery for the fusion protein immunofluorescence was entirely accounted for by protein synthesis. CONCLUSION: The covalent linkage between beta2AR and Galphas does not appear to alter the initial endocytic translocation of the two proteins, although there is reduced efficiency. It does, however, completely disrupt the process of receptor and G protein recycling. We conclude that the physical separation between receptor and Galpha is not necessary for the transit to early endosomes, but is an essential requirement for the correct post-endocytic sorting and recycling of the two proteins.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists , Cells, Cultured , Fluorescent Antibody Technique , GTP-Binding Protein alpha Subunits/genetics , Gene Expression Regulation , Humans , Models, Biological , Phosphorylation , Receptors, Adrenergic, beta-2/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
9.
Neuroreport ; 19(5): 531-5, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18388733

ABSTRACT

Che-1 is a nuclear protein involved in the regulation of gene transcription and cell proliferation. It has also been shown to localize to the cytoplasm of postmitotic neuronal cells, where it is able to interact with the microtubule-associated protein tau. Cyclin-dependent kinase 5 (Cdk5) is a postmitotic proline-directed serine/threonine kinase that hyperphosphorylates tau under pathological conditions. We observed that Che-1 overexpression induces Cdk5 expression both at the mRNA and protein levels. Furthermore, we show that Che-1 directly interacts with Cdk5 protein in vivo. Cdk5/Che-1 complex formation does not compete with Cdk5/p35 interaction, thus Che-1 is able to bind the active kinase complex. Finally, we demonstrated that Che-1 is itself a Cdk5 substrate.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Gene Expression Regulation , Gene Expression/physiology , Neurons/physiology , Transcription Factors/physiology , Animals , Animals, Newborn , Cells, Cultured , Cerebellum/cytology , Cyclin-Dependent Kinase 5/genetics , Gene Expression Regulation/genetics , Humans , Immunoprecipitation/methods , Mice , Nuclear Proteins , Rats , Rats, Wistar , Transcription Factors/genetics , Transfection/methods
10.
PLoS One ; 2(8): e774, 2007 Aug 22.
Article in English | MEDLINE | ID: mdl-17712422

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is a severe muscle degenerative disease, due to absence of dystrophin. There is currently no effective treatment for DMD. Our aim is to up-regulate the expression level of the dystrophin related gene utrophin in DMD, complementing in this way the lack of dystrophin functions. To this end we designed and engineered several synthetic zinc finger based transcription factors. In particular, we have previously shown that the artificial three zinc finger protein named Jazz, fused with the appropriate effector domain, is able to drive the transcription of a test gene from the utrophin promoter "A". Here we report on the characterization of Vp16-Jazz-transgenic mice that specifically over-express the utrophin gene at the muscular level. A Chromatin Immunoprecipitation assay (ChIP) demonstrated the effective access/binding of the Jazz protein to active chromatin in mouse muscle and Vp16-Jazz was shown to be able to up-regulate endogenous utrophin gene expression by immunohistochemistry, western blot analyses and real-time PCR. To our knowledge, this is the first example of a transgenic mouse expressing an artificial gene coding for a zinc finger based transcription factor. The achievement of Vp16-Jazz transgenic mice validates the strategy of transcriptional targeting of endogenous genes and could represent an exclusive animal model for use in drug discovery and therapeutics.


Subject(s)
Mice, Transgenic , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism , Up-Regulation , Utrophin/metabolism , Animals , Herpes Simplex Virus Protein Vmw65/genetics , Herpes Simplex Virus Protein Vmw65/metabolism , Humans , Mice , Microarray Analysis , Muscle Contraction/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Recombinant Fusion Proteins/genetics , Tissue Distribution , Transcription Factors/genetics , Utrophin/genetics , Zinc Fingers/genetics
11.
Biochem Cell Biol ; 85(4): 477-83, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17713582

ABSTRACT

Che-1/AATF (Che-1) was originally characterized as an interacting protein for RNA polymerase II. In addition to transcriptional regulation, the evidence suggests that Che-1 has a viral factor-like S phase promoting role in counteracting Rb repression to facilitate E2F-dependent transactivation during G1-S transition. Recently, Che-1 was found to play an important role in the DNA damage response and cell-cycle checkpoint control. Genetic studies in mice revealed that Che-1 is essential for preimplantation development and the establishment of embryonic gene expression. Importantly, several findings showed that Che-1 participates in inhibiting apoptotic process. Thus, Che-1 emerges as an important adaptor that connects transcriptional regulation, cell-cycle progression, checkpoint control, and apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Cell Cycle/physiology , Gene Expression Regulation , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/genetics , Genes, cdc , Humans , Leukemia/metabolism , Molecular Sequence Data , Repressor Proteins/genetics , Sequence Alignment , Transcription Factors/genetics
12.
J Biol Chem ; 282(27): 19685-91, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17468107

ABSTRACT

We have previously demonstrated that DNA damage leads to stabilization and accumulation of Che-1, an RNA polymerase II-binding protein that plays an important role in transcriptional activation of p53 and in maintenance of the G(2)/M checkpoint. Here we show that Che-1 is down-regulated during the apoptotic process. We found that the E3 ligase HMD2 physically and functionally interacts with Che-1 and promotes its degradation via the ubiquitin-dependent proteasomal system. Furthermore, we found that in response to apoptotic stimuli Che-1 interacts with the peptidyl-prolyl isomerase Pin1 and that conformational changes generated by Pin1 are required for Che-1/HDM2 interaction. Notably, a Che-1 mutant lacking the capacity to bind Pin1 exhibits an increased half-life and this correlates with a diminished apoptosis in response to genotoxic stress. Our results establish Che-1 as a new Pin1 and HDM2 target and confirm its important role in the cellular response to DNA damage.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , DNA Damage , Peptidylprolyl Isomerase/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Division , Cell Line, Tumor , Down-Regulation , G2 Phase , Humans , Mice , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
J Cell Sci ; 120(Pt 11): 1852-8, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17488777

ABSTRACT

Neurotrophin receptor-interacting MAGE homolog (NRAGE) has been recently identified as a cell-death inducer, involved in molecular events driving cells through apoptotic networks during neuronal development. Recently, we have focused on the functional role of Che-1, also known as apoptosis-antagonizing transcription factor (AATF), a protein involved in cell cycle control and gene transcription. Increasing evidence suggests that Che-1 is involved in apoptotic signalling in neural tissues. In cortical neurons Che-1 exhibits an anti-apoptotic activity, protecting cells from neuronal damage induced by amyloid beta-peptide. Here, we report that Che-1 interacts with NRAGE and that an EGFP-NRAGE fusion protein inhibits nuclear localization of Che-1, by sequestering it within the cytoplasmic compartment. Furthermore, NRAGE overexpression downregulates endogenous Che-1 by targeting it for proteasome-dependent degradation. Finally, we propose that Che-1 is a functional antagonist of NRAGE, because its overexpression completely reverts NRAGE-induced cell-death.


Subject(s)
Antigens, Neoplasm/metabolism , Apoptosis Regulatory Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Antigens, Neoplasm/chemistry , Apoptosis Regulatory Proteins/chemistry , Cell Death , Cell Nucleus/metabolism , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Neoplasm Proteins/chemistry , Protein Binding , Protein Transport , Repressor Proteins/chemistry , Transcription Factors/chemistry
14.
Cell Cycle ; 6(7): 804-6, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17377493

ABSTRACT

Che-1 is a RNA polymerase II binding protein involved in the transcriptional regulation of E2F target-genes and in cell proliferation. Recently, it has been shown that Che-1 accumulates in cells responding to genotoxic agents, such as Doxorubicin and ionizing radiations. The DNA damage-activated checkpoint kinases ATM and Chk2 interact with and phosphorylate Che-1, enhancing its accumulation and stability, and promoting Che-1-mediated transcription of p53-responsive genes and of p53 itself, as evidenced by microarray analysis. This transcriptional response is suppressed by expression of a Che-1 mutant lacking ATM and Chk2 phosphorylation amino acid residues, or by depletion of Che-1 by RNA silencing. In addition, chromatin immunoprecipitation analysis has shown that Che-1 is released from the E2F-target genes and recruited to the p21 and p53 promoters after DNA damage. Lastly, Che-1 contributes to the maintenance of the G2/M checkpoint in response to genotoxic stresses. These findings identify a new mechanism by which the checkpoint kinases regulate, via the novel effector Che-1, the p53 pathway.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Genes, cdc/physiology , Repressor Proteins/physiology , Signal Transduction/genetics , Transcription Factors/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , E2F Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Regulatory Elements, Transcriptional/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
15.
Cancer Cell ; 10(6): 473-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157788

ABSTRACT

Che-1 is a RNA polymerase II-binding protein involved in the transcription of E2F target genes and induction of cell proliferation. Here we show that Che-1 contributes to DNA damage response and that its depletion sensitizes cells to anticancer agents. The checkpoint kinases ATM/ATR and Chk2 interact with Che-1 and promote its phosphorylation and accumulation in response to DNA damage. These Che-1 modifications induce a specific recruitment of Che-1 on the TP53 and p21 promoters. Interestingly, it has a profound effect on the basal expression of p53, which is preserved following DNA damage. Notably, Che-1 contributes to the maintenance of the G2/M checkpoint induced by DNA damage. These findings identify a mechanism by which checkpoint kinases regulate responses to DNA damage.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Genes, p53 , Protein Serine-Threonine Kinases/physiology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/physiology , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Division , Checkpoint Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage , G2 Phase , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Promoter Regions, Genetic , Transcription, Genetic
16.
J Cell Sci ; 118(Pt 18): 4253-60, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16141233

ABSTRACT

Here, we show that the subcellular localization of alpha-like RNA polymerase II core subunit 3 (RPB3) is regulated during muscle differentiation. We have recently demonstrated that the expression of RPB3 is regulated during muscle differentiation and that, inside RNA polymerase II (RNAP II), it is directly involved in contacting regulatory proteins such as the myogenic transcription factor Myogenin and activating transcription factor ATF4. We show for the first time, that RPB3, in addition to its presence and role inside the RNAP II core enzyme, accumulates in the cytoplasm of cycling myogenic cells and migrates to the nucleus upon induction of the differentiation program. Furthermore, using human RPB3 as bait in a yeast two-hybrid system, we have isolated a novel RPB3 cytoplasmic interacting protein, HCR. HCR, previously identified as alpha-helix coiled-coil rod homologue, is one of the psoriasis vulgaris (PV) candidate genes. In cycling myogenic C2C7 cells, we show that the RPB3 protein directly interacts with HCR within the cytoplasm. Finally, knocking down HCR expression by RNA interference, we demonstrate that HCR acts as cytoplasmic docking site for RPB3.


Subject(s)
Carrier Proteins/metabolism , RNA Polymerase II/metabolism , Animals , Blotting, Western , Carrier Proteins/genetics , Cell Differentiation/physiology , Cytoplasm/enzymology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Myoblasts/cytology , Myoblasts/enzymology , Myoblasts/metabolism , NIH 3T3 Cells , Protein Structure, Tertiary , Psoriasis/genetics , RNA Interference , RNA Polymerase II/genetics
17.
J Biol Chem ; 278(38): 36496-504, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12847090

ABSTRACT

Che-1 is a recently identified human RNA polymerase II binding protein involved in the regulation of gene transcription and cell proliferation. We previously demonstrated that Che-1 inhibits the Rb growth-suppressing function by interfering with Rb-mediated HDAC1 recruitment on E2F target gene promoters. By hybridization of cancer profile arrays, we found that Che-1 expression is strongly down-regulated in several tumors, including colon and kidney carcinomas, compared with the relative normal tissues. Consistent with these data, Che-1 overexpression inhibits proliferation of HCT116 and LoVo human colon carcinoma cell lines by activation of the cyclin-dependent kinase inhibitor p21WAF1/Cip1 in a p53-independent manner and by promoting growth arrest at the G1 phase of the cell cycle. Che-1 activates p21WAF1/Cip1 by displacing histone deacetylase (HDAC)1 from the Sp1 binding sites of the p21WAF1/Cip1 gene promoter and accumulating acetylated histone H3 on these sites. Accordingly, Che-1-specific RNA interference negatively affects p21WAF1/Cip1 transactivation and increases cell proliferation in HCT116 cells. Taken together, our results indicate that Che-1 can be considered a general HDAC1 competitor and its down-regulation is involved in colon carcinoma cell proliferation.


Subject(s)
Colonic Neoplasms/pathology , Cyclins/genetics , Histone Deacetylases/metabolism , Promoter Regions, Genetic , Base Sequence , Binding, Competitive , Blotting, Western , Cell Division , Cell Line, Tumor , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Dose-Response Relationship, Drug , Down-Regulation , Glutathione Transferase/metabolism , Histone Deacetylase 1 , Histones/metabolism , Humans , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Precipitin Tests , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/metabolism , Time Factors , Tissue Distribution , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/metabolism
18.
FEBS Lett ; 547(1-3): 15-9, 2003 Jul 17.
Article in English | MEDLINE | ID: mdl-12860379

ABSTRACT

RPB3 is a core subunit of RNA polymerase II (pol II) that, together with the RPB11 subunit, forms the heterodimer considered as a functional counterpart of the bacterial alpha subunit homodimer involved in promoter recognition. We previously employed the yeast two-hybrid system and identified an interaction between RPB3 and the myogenic transcription factor myogenin, demonstrating an involvement of this subunit in muscle differentiation. In this paper we report the interaction between RPB3 and another known transcription factor, ATF4. We found that the intensity of the interaction between RPB3 and ATF4 is similar to the one between RPB3 and myogenin. This interaction involves an RPB3 specific region not homologous to the prokaryotic alpha subunit. We demonstrated that RBP3 is able to enhance ATF4 transactivation, whereas the region of RPB3 (Sud) that contacts ATF4, when used as a dominant negative, markedly inhibits ATF4 transactivation activity. Interestingly, ATF4 protein level, as reported for its partner RPB3, increases during C2C7 cell line muscle differentiation.


Subject(s)
Muscle, Skeletal/enzymology , Protein Subunits/metabolism , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Activating Transcription Factor 4 , Binding Sites , Cell Differentiation , Cloning, Molecular , Gene Library , Glutathione Transferase/metabolism , Humans , Muscle, Skeletal/cytology , Protein Subunits/genetics , RNA Polymerase II/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Sequence Deletion , Transcription Factors/genetics
19.
Cancer Cell ; 2(5): 387-99, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12450794

ABSTRACT

DNA tumor virus oncoproteins bind and inactivate Rb by interfering with the Rb/HDAC1 interaction. Che-1 is a recently identified human Rb binding protein that inhibits the Rb growth suppressing function. Here we show that Che-1 contacts the Rb pocket region and competes with HDAC1 for Rb binding site, removing HDAC1 from the Rb/E2F complex in vitro and from the E2F target promoters in vivo. Che-1 overexpression activates DNA synthesis in quiescent NIH-3T3 cells through HDAC1 displacement. Consistently, Che-1-specific RNA interference affects E2F activity and cell proliferation in human fibroblasts but not in the pocket protein-defective 293 cells. These findings indicate the existence of a pathway of Rb regulation supporting Che-1 as the cellular counterpart of DNA tumor virus oncoproteins.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Histone Deacetylases/metabolism , Repressor Proteins , Retinoblastoma Protein/physiology , 3T3 Cells , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Cell Division , Cell Line , Conserved Sequence , E2F Transcription Factors , Glutathione/metabolism , Histone Deacetylase 1 , Histone Deacetylases/genetics , Humans , Mice , Models, Biological , Mutation , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Sequence Alignment , Sequence Deletion , Transcription Factors/metabolism
20.
FASEB J ; 16(12): 1639-41, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12207009

ABSTRACT

RNA polymerase II core subunit 3 (RPB3) is an a-like core subunit of RNA polymerase II (pol II). It is selectively down-regulated upon treatment with doxorubicin (dox). Due to the failure of skeletal muscle cells to differentiate when exposed to dox, we hypothesized that RPB3 is involved in muscle differentiation. To this end, we have isolated human muscle RPB3-interacting proteins by using yeast two-hybrid screening. It is of interest that an interaction between RPB3 and the myogenic transcription factor myogenin was identified. This interaction involves a specific region of RPB3 protein that is not homologous to the prokaryotic a subunit. Although RPB3 contacts the basic helix-loop-helix (HLH) region of myogenin, it does not bind other HLH myogenic factors such as MyoD, Myf5, and MRF4. Coimmunoprecipitation experiments indicate that myogenin contacts the pol II complex and that the RPB3 subunit is responsible for this interaction. We show that RPB3 expression is regulated during muscle differentiation. Exogenous expression of RPB3 slightly promotes myogenin transactivation activity and muscle differentiation, whereas the region of RPB3 that contacts myogenin, when used as a dominant negative molecule (Sud), counteracts these effects. These results indicate for the first time that the RPB3 pol II subunit is involved in the regulation of tissue-specific transcription.


Subject(s)
Cell Differentiation/physiology , Myogenin/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins , Animals , Binding Sites , Blotting, Western , Cell Differentiation/genetics , Cell Line , Humans , Mice , Myocardium/cytology , Myocardium/metabolism , Myogenin/genetics , Protein Binding , RNA Polymerase II/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...