Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cancer Lett ; 589: 216820, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38574883

ABSTRACT

One in three Triple Negative Breast Cancer (TNBC) is Homologous Recombination Deficient (HRD) and susceptible to respond to PARP inhibitor (PARPi), however, resistance resulting from functional HR restoration is frequent. Thus, pharmacologic approaches that induce HRD are of interest. We investigated the effectiveness of CDK-inhibition to induce HRD and increase PARPi sensitivity of TNBC cell lines and PDX models. Two CDK-inhibitors (CDKi), the broad range dinaciclib and the CDK12-specific SR-4835, strongly reduced the expression of key HR genes and impaired HR functionality, as illustrated by BRCA1 and RAD51 nuclear foci obliteration. Consequently, both CDKis showed synergism with olaparib, as well as with cisplatin and gemcitabine, in a range of TNBC cell lines and particularly in olaparib-resistant models. In vivo assays on PDX validated the efficacy of dinaciclib which increased the sensitivity to olaparib of 5/6 models, including two olaparib-resistant and one BRCA1-WT model. However, no olaparib response improvement was observed in vivo with SR-4835. These data support that the implementation of CDK-inhibitors could be effective to sensitize TNBC to olaparib as well as possibly to cisplatin or gemcitabine.


Subject(s)
Antineoplastic Agents , Piperazines , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Cisplatin/pharmacology , Cisplatin/therapeutic use , Gemcitabine , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Phthalazines/pharmacology , Phthalazines/therapeutic use , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Line, Tumor
2.
Int J Mol Sci ; 23(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36012478

ABSTRACT

E4F1 is essential for early embryonic mouse development and for controlling the balance between proliferation and survival of actively dividing cells. We previously reported that E4F1 is essential for the survival of murine p53-deficient cancer cells by controlling the expression of genes involved in mitochondria functions and metabolism, and in cell-cycle checkpoints, including CHEK1, a major component of the DNA damage and replication stress responses. Here, combining ChIP-Seq and RNA-Seq approaches, we identified the transcriptional program directly controlled by E4F1 in Human Triple-Negative Breast Cancer cells (TNBC). E4F1 binds and regulates a limited list of direct target genes (57 genes) in these cells, including the human CHEK1 gene and, surprisingly, also two other genes encoding post-transcriptional regulators of the ATM/ATR-CHK1 axis, namely, the TTT complex component TTI2 and the phosphatase PPP5C, that are essential for the folding and stability, and the signaling of ATM/ATR kinases, respectively. Importantly, E4F1 also binds the promoter of these genes in vivo in Primary Derived Xenograft (PDX) of human TNBC. Consequently, the protein levels and signaling of CHK1 but also of ATM/ATR kinases are strongly downregulated in E4F1-depleted TNBC cells resulting in a deficiency of the DNA damage and replicative stress response in these cells. The E4F1-depleted cells fail to arrest into S-phase upon treatment with the replication-stalling agent Gemcitabine, and are highly sensitized to this drug, as well as to other DNA-damaging agents, such as Cisplatin. Altogether, our data indicate that in breast cancer cells the ATM/ATR-CHK1 signaling pathway and DNA damage-stress response are tightly controlled at the transcriptional and post-transcriptional level by E4F1.


Subject(s)
Repressor Proteins , Transcription Factors , Triple Negative Breast Neoplasms , Ubiquitin-Protein Ligases , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Mice , Phosphorylation , Protein Kinases/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Oncotarget ; 8(56): 95206-95222, 2017 Nov 10.
Article in English | MEDLINE | ID: mdl-29221122

ABSTRACT

Reduction in nucleotide pools through the inhibition of mitochondrial enzyme dihydroorotate dehydrogenase (DHODH) has been demonstrated to effectively reduce cancer cell proliferation and tumour growth. The current study sought to investigate whether this antiproliferative effect could be enhanced by combining Chk1 kinase inhibition. The pharmacological activity of DHODH inhibitor teriflunomide was more selective towards transformed mouse embryonic fibroblasts than their primary or immortalised counterparts, and this effect was amplified when cells were subsequently exposed to PF477736 Chk1 inhibitor. Flow cytometry analyses revealed substantial accumulations of cells in S and G2/M phases, followed by increased cytotoxicity which was characterised by caspase 3-dependent induction of cell death. Associating PF477736 with teriflunomide also significantly sensitised SUM159 and HCC1937 human triple negative breast cancer cell lines to dihydroorotate dehydrogenase inhibition. The main characteristic of this effect was the sustained accumulation of teriflunomide-induced DNA damage as cells displayed increased phospho serine 139 H2AX (γH2AX) levels and concentration-dependent phosphorylation of Chk1 on serine 345 upon exposure to the combination as compared with either inhibitor alone. Importantly a similar significant increase in cell death was observed upon dual siRNA mediated depletion of Chk1 and DHODH in both murine and human cancer cell models. Altogether these results suggest that combining DHODH and Chk1 inhibitions may be a strategy worth considering as a potential alternative to conventional chemotherapies.

4.
Proc Natl Acad Sci U S A ; 113(39): 11004-9, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27621431

ABSTRACT

The multifunctional protein E4 transcription factor 1 (E4F1) is an essential regulator of epidermal stem cell (ESC) maintenance. Here, we found that E4F1 transcriptionally regulates a metabolic program involved in pyruvate metabolism that is required to maintain skin homeostasis. E4F1 deficiency in basal keratinocytes resulted in deregulated expression of dihydrolipoamide acetyltransferase (Dlat), a gene encoding the E2 subunit of the mitochondrial pyruvate dehydrogenase (PDH) complex. Accordingly, E4f1 knock-out (KO) keratinocytes exhibited impaired PDH activity and a redirection of the glycolytic flux toward lactate production. The metabolic reprogramming of E4f1 KO keratinocytes associated with remodeling of their microenvironment and alterations of the basement membrane, led to ESC mislocalization and exhaustion of the ESC pool. ShRNA-mediated depletion of Dlat in primary keratinocytes recapitulated defects observed upon E4f1 inactivation, including increased lactate secretion, enhanced activity of extracellular matrix remodeling enzymes, and impaired clonogenic potential. Altogether, our data reveal a central role for Dlat in the metabolic program regulated by E4F1 in basal keratinocytes and illustrate the importance of PDH activity in skin homeostasis.


Subject(s)
DNA-Binding Proteins/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Homeostasis , Mitochondrial Proteins/metabolism , Skin/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Basement Membrane/metabolism , Cell Adhesion , Cells, Cultured , Cellular Microenvironment , DNA-Binding Proteins/deficiency , Dihydrolipoyllysine-Residue Acetyltransferase/genetics , Epidermal Cells , Epidermis/metabolism , Gene Expression Regulation , Keratinocytes/cytology , Keratinocytes/metabolism , Mice, Knockout , Mitochondrial Proteins/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Pyruvates/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins , Stem Cells/metabolism , Transcription Factors/deficiency , Ubiquitin-Protein Ligases
5.
Proc Natl Acad Sci U S A ; 113(39): 10998-1003, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27621446

ABSTRACT

The mitochondrial pyruvate dehydrogenase (PDH) complex (PDC) acts as a central metabolic node that mediates pyruvate oxidation and fuels the tricarboxylic acid cycle to meet energy demand. Here, we reveal another level of regulation of the pyruvate oxidation pathway in mammals implicating the E4 transcription factor 1 (E4F1). E4F1 controls a set of four genes [dihydrolipoamide acetlytransferase (Dlat), dihydrolipoyl dehydrogenase (Dld), mitochondrial pyruvate carrier 1 (Mpc1), and solute carrier family 25 member 19 (Slc25a19)] involved in pyruvate oxidation and reported to be individually mutated in human metabolic syndromes. E4F1 dysfunction results in 80% decrease of PDH activity and alterations of pyruvate metabolism. Genetic inactivation of murine E4f1 in striated muscles results in viable animals that show low muscle PDH activity, severe endurance defects, and chronic lactic acidemia, recapitulating some clinical symptoms described in PDC-deficient patients. These phenotypes were attenuated by pharmacological stimulation of PDH or by a ketogenic diet, two treatments used for PDH deficiencies. Taken together, these data identify E4F1 as a master regulator of the PDC.


Subject(s)
DNA-Binding Proteins/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , DNA-Binding Proteins/deficiency , Diet, Ketogenic , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscle Fibers, Skeletal/metabolism , Muscle, Striated/metabolism , Phenotype , Pyruvic Acid/metabolism , Repressor Proteins , Transcription Factors/deficiency , Ubiquitin-Protein Ligases
6.
Genom Data ; 5: 368-70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26484288

ABSTRACT

This Data in Brief report describes the experimental and bioinformatic procedures that we used to analyze and interpret E4F1 ChIP-seq experiments published in Rodier et al. (2015) [10]. Raw and processed data are available at the GEO DataSet repository under the subseries # GSE57228. E4F1 is a ubiquitously expressed zinc-finger protein of the GLI-Kruppel family that was first identified in the late eighties as a cellular transcription factor targeted by the adenoviral oncoprotein E1A13S (Ad type V) and required for the transcription of adenoviral genes (Raychaudhuri et al., 1987) [8]. It is a multifunctional factor that also acts as an atypical E3 ubiquitin ligase for p53 (Le Cam et al., 2006) [2]. Using KO mouse models we then demonstrated that E4F1 is essential for early embryonic development (Le Cam et al., 2004), for proliferation of mouse embryonic cell (Rodier et al., 2015), for the maintenance of epidermal stem cells (Lacroix et al., 2010) [6], and strikingly, for the survival of cancer cells (Hatchi et al., 2007) [4]; (Rodier et al., 2015) [10]. The latter survival phenotype was p53-independent and suggested that E4F1 was controlling a transcriptional program driving essential functions in cancer cells. To identify this program, we performed E4F1 ChIP-seq analyses in primary Mouse Embryonic Fibroblasts (MEF) and in p53(-/-), H-Ras(V12)-transformed MEFs. The program directly controlled by E4F1 was obtained by intersecting the lists of E4F1 genomic targets with the lists of genes differentially expressed in E4F1 KO and E4F1 WT cells (Rodier et al., 2015). We describe hereby how we improved our ChIP-seq analyses workflow by applying prefilters on raw data and by using a combination of two publicly available programs, Cisgenome and QESEQ.

7.
Cell Rep ; 11(2): 220-33, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25843721

ABSTRACT

Recent data support the notion that a group of key transcriptional regulators involved in tumorigenesis, including MYC, p53, E2F1, and BMI1, share an intriguing capacity to simultaneously regulate metabolism and cell cycle. Here, we show that another factor, the multifunctional protein E4F1, directly controls genes involved in mitochondria functions and cell-cycle checkpoints, including Chek1, a major component of the DNA damage response. Coordination of these cellular functions by E4F1 appears essential for the survival of p53-deficient transformed cells. Acute inactivation of E4F1 in these cells results in CHK1-dependent checkpoint deficiency and multiple mitochondrial dysfunctions that lead to increased ROS production, energy stress, and inhibition of de novo pyrimidine synthesis. This deadly cocktail leads to the accumulation of uncompensated oxidative damage to proteins and extensive DNA damage, ending in cell death. This supports the rationale of therapeutic strategies simultaneously targeting mitochondria and CHK1 for selective killing of p53-deficient cancer cells.


Subject(s)
DNA-Binding Proteins/genetics , Mitochondria/metabolism , Neoplasms/genetics , Protein Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Survival , Checkpoint Kinase 1 , DNA Damage/genetics , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Mice , Mitochondria/pathology , Mouse Embryonic Stem Cells/metabolism , Neoplasms/metabolism , Protein Kinases/biosynthesis , Pyrimidines/biosynthesis , Repressor Proteins , Stress, Physiological/genetics , Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Ubiquitin-Protein Ligases
8.
Autophagy ; 7(12): 1566-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22024746

ABSTRACT

The multifunctional E4F1 protein was originally identified as a cellular target of the E1A adenoviral oncoprotein. Although E4F1 is implicated in several key oncogenic pathways, its roles in tumorigenesis remain unclear. Using a genetically engineered mouse model of myeloid leukemia (histiocytic sarcomas, HS) based on the genetic inactivation of the tumor suppressor Ink4a/Arf locus, we have recently unraveled an unsuspected function of E4F1 in the survival of leukemic cells. In vivo, genetic ablation of E4F1 in established myeloid tumors results in tumor regression. E4F1 inactivation results in a cascade of alterations originating from dysfunctional mitochondria that induce increased reactive oxygen species (ROS) levels and ends in massive autophagic cell death in HS transformed, but not normal myeloid cells. E4F1 depletion also induces cell death in various human myeloid leukemic cell lines, including acute myeloid leukemic (AML) cell lines. Interestingly, the E4F1 protein is overexpressed in a large proportion of human AML samples. These data provide new insights into E4F1-associated survival functions implicated in tumorigenesis and could open the path for new therapeutic strategies.


Subject(s)
Autophagy , Leukemia, Myeloid/pathology , Repressor Proteins/metabolism , Animals , Cell Survival , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Humans , Leukemia, Myeloid/metabolism , Mice , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
9.
J Exp Med ; 208(7): 1403-17, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21708927

ABSTRACT

The multifunctional E4F1 protein was originally discovered as a target of the E1A viral oncoprotein. Growing evidence indicates that E4F1 is involved in key signaling pathways commonly deregulated during cell transformation. In this study, we investigate the influence of E4F1 on tumorigenesis. Wild-type mice injected with fetal liver cells from mice lacking CDKN2A, the gene encoding Ink4a/Arf, developed histiocytic sarcomas (HSs), a tumor originating from the monocytic/macrophagic lineage. Cre-mediated deletion of E4F1 resulted in the death of HS cells and tumor regression in vivo and extended the lifespan of recipient animals. In murine and human HS cell lines, E4F1 inactivation resulted in mitochondrial defects and increased production of reactive oxygen species (ROS) that triggered massive cell death. Notably, these defects of E4F1 depletion were observed in HS cells but not healthy primary macrophages. Short hairpin RNA-mediated depletion of E4F1 induced mitochondrial defects and ROS-mediated death in several human myeloid leukemia cell lines. E4F1 protein is overexpressed in a large subset of human acute myeloid leukemia samples. Together, these data reveal a role for E4F1 in the survival of myeloid leukemic cells and support the notion that targeting E4F1 activities might have therapeutic interest.


Subject(s)
DNA-Binding Proteins/deficiency , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Repressor Proteins/deficiency , Transcription Factors/deficiency , Animals , Autophagy/physiology , Base Sequence , Cell Death/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/metabolism , Histiocytic Sarcoma/pathology , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Oxidative Stress , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Ubiquitin-Protein Ligases
10.
Proc Natl Acad Sci U S A ; 107(49): 21076-81, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21088222

ABSTRACT

A growing body of evidence suggests that the multifunctional protein E4F1 is involved in signaling pathways that play essential roles during normal development and tumorigenesis. We generated E4F1 conditional knockout mice to address E4F1 functions in vivo in newborn and adult skin. E4F1 inactivation in the entire skin or in the basal compartment of the epidermis induces skin homeostasis defects, as evidenced by transient hyperplasia in the interfollicular epithelium and alteration of keratinocyte differentiation, followed by loss of cellularity in the epidermis and severe skin ulcerations. E4F1 depletion alters clonogenic activity of epidermal stem cells (ESCs) ex vivo and ends in exhaustion of the ESC pool in vivo, indicating that the lesions observed in the E4F1 mutant skin result, at least in part, from cell-autonomous alterations in ESC maintenance. The clonogenic potential of E4F1 KO ESCs is rescued by Bmi1 overexpression or by Ink4a/Arf or p53 depletion. Skin phenotype of E4F1 KO mice is also delayed in animals with Ink4a/Arf and E4F1 compound gene deficiencies. Our data identify a regulatory axis essential for ESC-dependent skin homeostasis implicating E4F1 and the Bmi1-Arf-p53 pathway.


Subject(s)
DNA-Binding Proteins/physiology , Epidermal Cells , Homeostasis , Stem Cells/physiology , Transcription Factors/physiology , Age Factors , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Phenotype , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases
11.
Mol Cancer Ther ; 9(6): 1740-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20530718

ABSTRACT

Increased de novo fatty acid (FA) synthesis is one hallmark of tumor cells, including prostate cancer. We present here our most recent results showing that lipid composition in human prostate cancer is characterized by an increased ratio of monounsaturated FA to saturated FA, compared with normal prostate, and evidence the overexpression of the lipogenic enzyme stearoyl-CoA desaturase 1 (SCD1) in human prostate cancer. As a new therapeutic strategy, we show that pharmacologic inhibition of SCD1 activity impairs lipid synthesis and results in decreased proliferation of both androgen-sensitive and androgen-resistant prostate cancer cells, abrogates the growth of prostate tumor xenografts in nude mice, and confers therapeutic benefit on animal survival. We show that these changes in lipid synthesis are translated into the inhibition of the AKT pathway and that the decrease in concentration of phosphatidylinositol-3,4,5-trisphosphate might at least partially mediate this effect. Inhibition of SCD1 also promotes the activation of AMP-activated kinase and glycogen synthase kinase 3alpha/beta, the latter on being consistent with a decrease in beta-catenin activity and mRNA levels of various beta-catenin growth-promoting transcriptional targets. Furthermore, we show that SCD1 activity is required for cell transformation by Ras oncogene. Together, our data support for the first time the concept of targeting the lipogenic enzyme SCD1 as a new promising therapeutic approach to block oncogenesis and prostate cancer progression.


Subject(s)
Disease Progression , Lipogenesis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids, Monounsaturated/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Lipogenesis/drug effects , Male , Mice , Stearoyl-CoA Desaturase/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
12.
Biochem J ; 428(1): 103-11, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20236090

ABSTRACT

ERK3 (extracellular-signal-regulated kinase 3) is an atypical MAPK (mitogen-activated protein kinase) that is suggested to play a role in cell-cycle progression and cellular differentiation. However, it is not known whether the function of ERK3 is regulated during the cell cycle. In the present paper, we report that ERK3 is stoichiometrically hyperphosphorylated during entry into mitosis and is dephosphorylated at the M-->G1 transition. The phosphorylation of ERK3 is associated with the accumulation of the protein in mitosis. In vitro phosphorylation of a series of ERK3-deletion mutants by mitotic cell extracts revealed that phosphorylation is confined to the unique C-terminal extension of the protein. Using MS analysis, we identified four novel phosphorylation sites, Ser684, Ser688, Thr698 and Ser705, located at the extreme C-terminus of ERK3. All four sites are followed by a proline residue. We have shown that purified cyclin B-Cdk1 (cyclindependent kinase 1) phosphorylates these sites in vitro and demonstrate that Cdk1 acts as a major Thr698 kinase in vivo. Reciprocally, we found that the phosphatases Cdc14A and Cdc14B (Cdc is cell-division cycle) bind to ERK3 and reverse its C-terminal phosphorylation in mitosis. Importantly, alanine substitution of the four C-terminal phosphorylation sites markedly decreased the half-life of ERK3 in mitosis, thereby linking phosphorylation to the stabilization of the kinase. The results of the present study identify a novel regulatory mechanism of ERK3 that operates in a cell-cycle-dependent manner.


Subject(s)
CDC2 Protein Kinase/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Mitosis , Phosphoprotein Phosphatases/metabolism , HeLa Cells , Humans , Phosphorylation , Transfection
13.
Cell Cycle ; 9(5): 975-9, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20160482

ABSTRACT

Skp2 is the substrate binding subunit of the SCF(Skp2) ubiquitin ligase, which plays a key role in the regulation of cell cycle progression. The activity of Skp2 is regulated by the APC(Cdh1), which targets Skp2 for degradation in early G(1) and prevent premature S phase entry. Overexpression of Skp2 leads to dysregulation of the cell cycle and is commonly observed in human cancers. We have previously shown that Skp2 is phosphorylated on Ser64 and Ser72 in vivo, and that these modifications regulate its stability. Recently, two studies have proposed a role for Ser72 phosphorylation in the cytosolic relocalization of Skp2 and in the assembly and activity of SCF(Skp2) ubiquitin ligase complex. We have revisited this question and analyzed the impact of Ser72 phosphorylation site mutations on the biological activity and subcellular localization of Skp2. We show here that phosphorylation of Ser72 does not control Skp2 binding to Skp1 and Cul1, has no influence on SCF(Skp2) ubiquitin ligase activity, and does not affect the subcellular localization of Skp2 in a panel of cell lines.


Subject(s)
S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Cullin Proteins/metabolism , G1 Phase , Humans , Phosphorylation , S Phase , S-Phase Kinase-Associated Proteins/analysis , Serine/metabolism
15.
J Cell Physiol ; 217(3): 778-88, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18720373

ABSTRACT

Mitogen-activated protein (MAP) kinases are typical examples of protein kinases whose enzymatic activity is mainly controlled by activation loop phosphorylation. The classical MAP kinases ERK1/ERK2, JNK, p38 and ERK5 all contain the conserved Thr-Xxx-Tyr motif in their activation loop that is dually phosphorylated by members of the MAP kinase kinases family. Much less is known about the regulation of the atypical MAP kinases ERK3 and ERK4. These kinases display structural features that distinguish them from other MAP kinases, notably the presence of a single phospho-acceptor site (Ser-Glu-Gly) in the activation loop. Here, we show that ERK3 and ERK4 are phosphorylated in their activation loop in vivo. This phosphorylation is exerted, at least in part, in trans by an upstream cellular kinase. Contrary to classical MAP kinases, activation loop phosphorylation of ERK3 and ERK4 is detected in resting cells and is not further stimulated by strong mitogenic or stress stimuli. However, phosphorylation can be modulated indirectly by interaction with the substrate MAP kinase-activated protein kinase 5 (MK5). Importantly, we found that activation loop phosphorylation of ERK3 and ERK4 stimulates their intrinsic catalytic activity and is required for the formation of stable active complexes with MK5 and, consequently, for efficient cytoplasmic redistribution of ERK3/ERK4-MK5 complexes. Our results demonstrate the importance of activation loop phosphorylation in the regulation of ERK3/ERK4 function and highlight differences in the regulation of atypical MAP kinases as compared to classical family members.


Subject(s)
Cytoplasm/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Nucleus/enzymology , Enzyme Activation , Humans , Mice , Models, Biological , NIH 3T3 Cells , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Transport , Rats , Substrate Specificity
16.
EMBO Rep ; 9(5): 452-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18404153

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) targets nuclear and cytoplasmic proteins. Here, we identified a nuclear protein, called cooperator of PRMT5 (COPR5), involved in the nuclear functions of PRMT5. COPR5 tightly binds to PRMT5, both in vitro and in living cells, but not to other members of the PRMT family. PRMT5 bound to COPR5 methylates histone H4 (R3) preferentially when compared with histone H3 (R8), suggesting that COPR5 modulates the substrate specificity of nuclear PRMT5-containing complexes, at least towards histones. Markedly, recombinant COPR5 binds to the amino terminus of histone H4 and is required to recruit PRMT5 to reconstituted nucleosomes in vitro. Consistently, COPR5 depletion in cells strongly reduces PRMT5 recruitment on chromatin at the PRMT5 target gene cyclin E1 (CCNE1) in vivo. Moreover, both COPR5 depletion and overexpression affect CCNE1 promoter expression. We propose that COPR5 is an important chromatin adaptor for PRMT5 to function on a subset of its target genes.


Subject(s)
Arginine/metabolism , Carrier Proteins/physiology , Histones/metabolism , Nuclear Proteins/metabolism , Protein Methyltransferases/physiology , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chromatin/metabolism , DNA Methylation , Genes, Reporter , Glutathione Transferase/metabolism , Humans , Luciferases/metabolism , Molecular Sequence Data , Nuclear Proteins/physiology , Protein Binding , Protein Methyltransferases/metabolism , Protein Structure, Secondary , Recombinant Proteins/metabolism
17.
EMBO J ; 27(4): 679-91, 2008 Feb 20.
Article in English | MEDLINE | ID: mdl-18239684

ABSTRACT

The p27(Kip1) ubiquitin ligase receptor Skp2 is often overexpressed in human tumours and displays oncogenic properties. The activity of SCF(Skp2) is regulated by the APC(Cdh1), which targets Skp2 for degradation. Here we show that Skp2 phosphorylation on Ser64/Ser72 positively regulates its function in vivo. Phosphorylation of Ser64, and to a lesser extent Ser72, stabilizes Skp2 by interfering with its association with Cdh1, without affecting intrinsic ligase activity. Cyclin-dependent kinase (CDK)2-mediated phosphorylation of Skp2 on Ser64 allows its expression in mid-G1 phase, even in the presence of active APC(Cdh1). Reciprocally, dephosphorylation of Skp2 by the mitotic phosphatase Cdc14B at the M --> G1 transition promotes its degradation by APC(Cdh1). Importantly, lowering the levels of Cdc14B accelerates cell cycle progression from mitosis to S phase in an Skp2-dependent manner, demonstrating epistatic relationship of Cdc14B and Skp2 in the regulation of G1 length. Thus, our results reveal that reversible phosphorylation plays a key role in the timing of Skp2 expression in the cell cycle.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Dual-Specificity Phosphatases/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Line , G1 Phase , HeLa Cells , Humans , Mice , Mutation , NIH 3T3 Cells , Phosphorylation , Rats , S-Phase Kinase-Associated Proteins/genetics , Transfection
18.
J Immunol ; 177(8): 5059-67, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015689

ABSTRACT

Activation of the innate arm of the immune system following pathogen infection relies on the recruitment of latent transcription factors involved in the induction of a subset of genes responsible for viral clearance. One of these transcription factors, IFN regulatory factor 3 (IRF-3), is targeted for proteosomal degradation following virus infection. However, the molecular mechanisms involved in this process are still unknown. In this study, we show that polyubiquitination of IRF-3 increases in response to Sendai virus infection. Using an E1 temperature-sensitive cell line, we demonstrate that polyubiquitination is required for the observed degradation of IRF-3. Inactivation of NEDD8-activating E1 enzyme also results in stabilization of IRF-3 suggesting the NEDDylation also plays a role in IRF-3 degradation following Sendai virus infection. In agreement with this observation, IRF-3 is recruited to Cullin1 following virus infection and overexpression of a dominant-negative mutant of Cullin1 significantly inhibits the degradation of IRF-3 observed in infected cells. We also asked whether the C-terminal cluster of phosphoacceptor sites of IRF-3 could serve as a destabilization signal and we therefore measured the half-life of C-terminal phosphomimetic IRF-3 mutants. Interestingly, we found them to be short-lived in contrast to wild-type IRF-3. In addition, no degradation of IRF-3 was observed in TBK1(-/-) mouse embryonic fibroblasts. All together, these data demonstrate that virus infection stimulates a host cell signaling pathway that modulates the expression level of IRF-3 through its C-terminal phosphorylation by the IkappaB kinase-related kinases followed by its polyubiquitination, which is mediated in part by a Cullin-based ubiquitin ligase.


Subject(s)
Cullin Proteins/metabolism , Interferon Regulatory Factor-3/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Diseases/metabolism , Animals , Cell Line , Humans , I-kappa B Kinase , Mice , Mice, Knockout , Phosphorylation , Sendai virus , Signal Transduction , Ubiquitin/metabolism
19.
J Cell Biol ; 168(1): 55-66, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15631990

ABSTRACT

Cell cycle progression is negatively regulated by the pocket proteins pRb, p107, and p130. However, the mechanisms responsible for this inhibition are not fully understood. Here, we show that overexpression of p107 in fibroblasts inhibits Cdk2 activation and delays S phase entry. The inhibition of Cdk2 activity is correlated with the accumulation of p27, consequent to a decreased degradation of the protein, with no change of Thr187 phosphorylation. Instead, we observed a marked decrease in the abundance of the F-box receptor Skp2 in p107-overexpressing cells. Reciprocally, Skp2 accumulates to higher levels in p107-/- embryonic fibroblasts. Ectopic expression of Skp2 restores p27 down-regulation and DNA synthesis to the levels observed in parental cells, whereas inactivation of Skp2 abrogates the inhibitory effect of p107 on S phase entry. We further show that the serum-dependent increase in Skp2 half-life observed during G1 progression is impaired in cells overexpressing p107. We propose that p107, in addition to its interaction with E2F, inhibits cell proliferation through the control of Skp2 expression and the resulting stabilization of p27.


Subject(s)
Cell Cycle Proteins/metabolism , G1 Phase/physiology , Nuclear Proteins/metabolism , S Phase/physiology , Animals , CDC2-CDC28 Kinases/metabolism , Carbohydrate Dehydrogenases/metabolism , Cell Cycle Proteins/genetics , Cell Line , Cullin Proteins/metabolism , Culture Media, Serum-Free , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mice , Mice, Knockout , Nuclear Proteins/genetics , RNA, Small Interfering/metabolism , Rats , Retinoblastoma-Like Protein p107 , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Mol Cell Biol ; 24(14): 6140-50, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15226418

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3) is an unstable mitogen-activated protein kinase homologue that is constitutively degraded by the ubiquitin-proteasome pathway in proliferating cells. Here we show that a lysineless mutant of ERK3 is still ubiquitinated in vivo and requires a functional ubiquitin conjugation pathway for its degradation. Addition of N-terminal sequence tags of increasing size stabilizes ERK3 by preventing its ubiquitination. Importantly, we identified a fusion peptide between the N-terminal methionine of ERK3 and the C-terminal glycine of ubiquitin in vivo by tandem mass spectrometry analysis. These findings demonstrate that ERK3 is conjugated to ubiquitin via its free NH(2) terminus. We found that large N-terminal tags also stabilize the expression of the cell cycle inhibitor p21 but not that of substrates ubiquitinated on internal lysine residues. Consistent with this observation, lysineless p21 is ubiquitinated and degraded in a ubiquitin-dependent manner in intact cells. Our results suggests that N-terminal ubiquitination is a more prevalent modification than originally recognized.


Subject(s)
Cyclins/metabolism , Cysteine Endopeptidases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/metabolism , Ubiquitin/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Stability , Humans , Lysine/metabolism , Mass Spectrometry , Mitogen-Activated Protein Kinase 6 , Mitogen-Activated Protein Kinases/genetics , Mutation , Peptides/genetics , Peptides/metabolism , Proteasome Endopeptidase Complex , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Tagged Sites
SELECTION OF CITATIONS
SEARCH DETAIL
...