Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1191908, 2023.
Article in English | MEDLINE | ID: mdl-37435061

ABSTRACT

Introduction: AATF/Che-1 over-expression in different tumors is well known and its effect on tumorigenicity is mainly due to its central role demonstrated in the oncogenic pathways of solid tumors, where it controls proliferation and viability. The effect exerted by tumors overexpressing Che-1 on the immune response has not yet been investigated. Methods: Starting from ChIP-sequencing data we confirmed Che-1 enrichment on Nectin-1 promoter. Several co-cultures experiments between NK-cells and tumor cells transduced by lentiviral vectors carrying Che-1-interfering sequence, analyzed by flow-cytometry have allowed a detailed characterization of NK receptors and tumor ligands expression. Results: Here, we show that Che-1 is able to modulate the expression of Nectin-1 ligand at the transcriptional level, leading to the impairment of killing activity of NK-cells. Nectin-1 down-modulation induces a modification in NK-cell ligands expression able to interact with activating receptors and to stimulate NK-cell function. In addition, NK-cells from Che-1 transgenic mice, confirming a reduced expression of activating receptors, exhibit impaired activation and a preferential immature status. Discussion: The critical equilibrium between NK-cell ligand expression on tumor cells and the interaction with NK cell receptors is affected by Che-1 over-expression and partially restored by Che-1 interference. The evidence of a new role for Che-1 as regulator of anti-tumor immunity supports the necessity to develop approaches able to target this molecule which shows a dual tumorigenic function as cancer promoter and immune response modulator.


Subject(s)
Carrier Proteins , Neoplasms , Animals , Mice , Ligands , Mice, Transgenic , Nectins/genetics , Neoplasms/genetics , RNA Polymerase II
2.
J Exp Clin Cancer Res ; 40(1): 232, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34266450

ABSTRACT

BACKGROUND: Che-1/AATF (Che-1) is an RNA polymerase II binding protein involved in several cellular processes, including proliferation, apoptosis and response to stress. We have recently demonstrated that Che-1 is able to promote cell proliferation by sustaining global histone acetylation in multiple myeloma (MM) cells where it interacts with histone proteins and competes with HDAC class I members for binding. METHODS: Site-directed Mutagenesis was performed to generate a Che-1 mutant (Che-1 3S) lacking three serine residues (Ser316, Ser320 and Ser321) in 308-325 aa region. Western blot experiments were conducted to examine the effect of depletion or over-expression of Che-1 and Che-1 3S mutant on histone acetylation, in different human cancer cell lines. Proliferation assays were assessed to estimate the change in cells number when Che-1 was over-expressed or deleted. Immunoprecipitation assays were performed to evaluate Che-1/histone H3 interaction when Ser316, Ser320 and Ser321 were removed. The involvement of CK2 kinase in Che-1 phosphorylation at these residues was analysed by in vitro kinase, 2D gel electrophoresis assays and mass spectrometry analysis. RESULTS: Here, we confirmed that Che-1 depletion reduces cell proliferation with a concomitant general histone deacetylation in several tumor cell lines. Furthermore, we provided evidence that CK2 protein kinase phosphorylates Che-1 at Ser316, Ser320 and Ser321 and that these modifications are required for Che-1/histone H3 binding. These results improve our understanding onto the mechanisms by which Che-1 regulates histone acetylation and cell proliferation. CONCLUSIONS: Che-1 phosphorylation at Ser316, Ser320 and Ser321 by CK2 promotes the interaction with histone H3 and represents an essential requirement for Che-1 pro-proliferative ability.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Casein Kinase II/metabolism , Repressor Proteins/metabolism , Cell Proliferation/physiology , Humans , Phosphorylation , Transfection
3.
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
4.
Nucleic Acids Res ; 48(11): 5891-5906, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32421830

ABSTRACT

Originally identified as an RNA polymerase II interactor, Che-1/AATF (Che-1) has now been recognized as a multifunctional protein involved in cell-cycle regulation and cancer progression, as well as apoptosis inhibition and response to stress. This protein displays a peculiar nucleolar localization and it has recently been implicated in pre-rRNA processing and ribosome biogenesis. Here, we report the identification of a novel function of Che-1 in the regulation of ribosomal RNA (rRNA) synthesis, in both cancer and normal cells. We demonstrate that Che-1 interacts with RNA polymerase I and nucleolar upstream binding factor (UBF) and promotes RNA polymerase I-dependent transcription. Furthermore, this protein binds to the rRNA gene (rDNA) promoter and modulates its epigenetic state by contrasting the recruitment of HDAC1. Che-1 downregulation affects RNA polymerase I and UBF recruitment on rDNA and leads to reducing rDNA promoter activity and 47S pre-rRNA production. Interestingly, Che-1 depletion induces abnormal nucleolar morphology associated with re-distribution of nucleolar proteins. Finally, we show that upon DNA damage Che-1 re-localizes from rDNA to TP53 gene promoter to induce cell-cycle arrest. This previously uncharacterized function of Che-1 confirms the important role of this protein in the regulation of ribosome biogenesis, cellular proliferation and response to stress.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , DNA, Ribosomal/genetics , Genes, rRNA/genetics , RNA Polymerase I/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints , Cell Line , Cell Nucleolus/metabolism , Cell Nucleolus/pathology , DNA Damage , DNA, Ribosomal/metabolism , Homeostasis , Humans , Phosphorylation , Pol1 Transcription Initiation Complex Proteins/metabolism , Promoter Regions, Genetic , Repressor Proteins/deficiency , Repressor Proteins/genetics , Ribosomes/metabolism
5.
EMBO Rep ; 19(3)2018 03.
Article in English | MEDLINE | ID: mdl-29367285

ABSTRACT

Despite progress in treating B-cell precursor acute lymphoblastic leukemia (BCP-ALL), disease recurrence remains the main cause of treatment failure. New strategies to improve therapeutic outcomes are needed, particularly in high-risk relapsed patients. Che-1/AATF (Che-1) is an RNA polymerase II-binding protein involved in proliferation and tumor survival, but its role in hematological malignancies has not been clarified. Here, we show that Che-1 is overexpressed in pediatric BCP-ALL during disease onset and at relapse, and that its depletion inhibits the proliferation of BCP-ALL cells. Furthermore, we report that c-Myc regulates Che-1 expression by direct binding to its promoter and describe a strict correlation between Che-1 expression and c-Myc expression. RNA-seq analyses upon Che-1 or c-Myc depletion reveal a strong overlap of the respective controlled pathways. Genomewide ChIP-seq experiments suggest that Che-1 acts as a downstream effector of c-Myc. These results identify the pivotal role of Che-1 in the control of BCP-ALL proliferation and present the protein as a possible therapeutic target in children with relapsed BCP-ALL.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , High-Throughput Nucleotide Sequencing , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Promoter Regions, Genetic/genetics
6.
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
7.
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
8.
Front Oncol ; 6: 34, 2016.
Article in English | MEDLINE | ID: mdl-26913241

ABSTRACT

The p53 protein is a key player in a wide range of protein networks that allow the state of "good health" of the cell. Not surprisingly, mutations of the TP53 gene are one of the most common alterations associated to cancer cells. Mutated forms of p53 (mtp53) not only lose the ability to protect the integrity of the genetic heritage of the cell but also acquire pro-oncogenic functions, behaving like dangerous accelerators of transformation and tumor progression. In recent years, many studies focused on investigating possible strategies aiming to counteract this mutant p53 "gain of function" but the results have not always been satisfactory. Che-1/AATF is a nuclear protein that binds to RNA polymerase II and plays a role in multiple fundamental processes, including control of transcription, cell cycle regulation, DNA damage response, and apoptosis. Several studies showed Che-1/AATF as an important endogenous regulator of p53 expression and activity in a variety of biological processes. Notably, this same regulation was more recently observed also on mtp53. The depletion of Che-1/AATF strongly reduces the expression of mutant p53 in several tumors in vitro and in vivo, making the cells an easier target for chemotherapy treatments. In this mini review, we report an overview of Che-1/AATF functions and discuss a possible role of Che-1/AATF in cancer therapy, with particular regard to its action on p53/mtp53.

9.
Front Genet ; 6: 141, 2015.
Article in English | MEDLINE | ID: mdl-25914721

ABSTRACT

The transcriptional cofactor Che-1/AATF is currently emerging as an important component of the DNA damage response (DDR) machinery, the complex signaling network that maintains genome integrity and prevents tumorigenesis. Moreover this protein is involved in a wide range of cellular pathways, regulating proliferation and survival in both physiological and pathological conditions. Notably, some evidence indicates that dysregulation of Che-1/AATF levels are associated with the transformation process and elevated levels of Che-1/AATF are required for tumor cell survival. It is for these reasons that Che-1/AATF has been regarded as an attractive, still theoretical, therapeutic target for cancer treatments. In this review, we will provide an updated overview of Che-1/AATF activities, from transcriptional regulation to DDR.

10.
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
11.
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
12.
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
13.
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
14.
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
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.
Dev Cell ; 6(5): 673-84, 2004 May.
Article in English | MEDLINE | ID: mdl-15130492

ABSTRACT

Fusion of undifferentiated myoblasts into multinucleated myotubes is a prerequisite for developmental myogenesis and postnatal muscle growth. We report that deacetylase inhibitors favor the recruitment and fusion of myoblasts into preformed myotubes. Muscle-restricted expression of follistatin is induced by deacetylase inhibitors and mediates myoblast recruitment and fusion into myotubes through a pathway distinct from those utilized by either IGF-1 or IL-4. Blockade of follistatin expression by RNAi-mediated knockdown, functional inactivation with either neutralizing antibodies or the antagonist protein myostatin, render myoblasts refractory to HDAC inhibitors. Muscles from animals treated with the HDAC inhibitor trichostatin A display increased production of follistatin and enhanced expression of markers of regeneration following muscle injury. These data identify follistatin as a central mediator of the fusigenic effects exerted by deacetylase inhibitors on skeletal muscles and establish a rationale for their use to manipulate skeletal myogenesis and promote muscle regeneration.


Subject(s)
Follistatin/antagonists & inhibitors , Histone Deacetylase Inhibitors , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/growth & development , Myoblasts, Skeletal/enzymology , Nuclear Proteins , Animals , Antibodies/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cyclic AMP Response Element-Binding Protein , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Follistatin/genetics , Follistatin/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Membrane Fusion/genetics , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , MyoD Protein/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , NFATC Transcription Factors , NIH 3T3 Cells , RNA Interference , Regeneration/drug effects , Regeneration/genetics , Transcription Factors/metabolism
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.
Mol Cell ; 12(1): 51-62, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12887892

ABSTRACT

Sir2 is a NAD(+)-dependent histone deacetylase that controls gene silencing, cell cycle, DNA damage repair, and life span. Prompted by the observation that the [NAD(+)]/[NADH] ratio is subjected to dynamic fluctuations in skeletal muscle, we have tested whether Sir2 regulates muscle gene expression and differentiation. Sir2 forms a complex with the acetyltransferase PCAF and MyoD and, when overexpressed, retards muscle differentiation. Conversely, cells with decreased Sir2 differentiate prematurely. To inhibit myogenesis, Sir2 requires its NAD(+)-dependent deacetylase activity. The [NAD(+)]/[NADH] ratio decreases as muscle cells differentiate, while an increased [NAD(+)]/[NADH] ratio inhibits muscle gene expression. Cells with reduced Sir2 levels are less sensitive to the inhibition imposed by an elevated [NAD(+)]/[NADH] ratio. These results indicate that Sir2 regulates muscle gene expression and differentiation by possibly functioning as a redox sensor. In response to exercise, food intake, and starvation, Sir2 may sense modifications of the redox state and promptly modulate gene expression.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/embryology , Muscle, Skeletal/enzymology , Sirtuins/metabolism , Acetyltransferases/metabolism , Animals , Cell Line , Histone Acetyltransferases , Humans , Mice , MyoD Protein/metabolism , NAD/metabolism , Oxidation-Reduction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sirtuin 2 , Sirtuins/genetics
19.
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
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
...