1
|
Javary J, Goupil E, Soulez M, Kanshin E, Bouchard A, Seternes OM, Thibault P, Labbé JC, Meloche S. Phosphoproteomic analysis identifies supervillin as an ERK3 substrate regulating cytokinesis and cell ploidy. J Cell Physiol 2024; 239:e30938. [PMID: 36576983 DOI: 10.1002/jcp.30938] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022]
Abstract
Extracellular signal-regulated kinase 3 (ERK3) is a poorly characterized member of the mitogen-activated protein (MAP) kinase family. Functional analysis of the ERK3 signaling pathway has been hampered by a lack of knowledge about the substrates and downstream effectors of the kinase. Here, we used large-scale quantitative phosphoproteomics and targeted gene silencing to identify direct ERK3 substrates and gain insight into its cellular functions. Detailed validation of one candidate substrate identified the gelsolin/villin family member supervillin (SVIL) as a bona fide ERK3 substrate. We show that ERK3 phosphorylates SVIL on Ser245 to regulate myosin II activation and cytokinesis completion in dividing cells. Depletion of SVIL or ERK3 leads to increased cytokinesis failure and multinucleation, a phenotype rescued by wild type SVIL but not by the non-phosphorylatable S245A mutant. Our results unveil a new function of the atypical MAP kinase ERK3 in cell division and the regulation of cell ploidy.
Collapse
Affiliation(s)
- Joaquim Javary
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Eugénie Goupil
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Mathilde Soulez
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Evgeny Kanshin
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
- NYU Langone Health, New York City, New York, USA
| | - Antoine Bouchard
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
| | | | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
- Department of Chemistry, Faculty of Arts and Sciences, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Claude Labbé
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Guégan JP, Lapouge M, Voisin L, Saba-El-Leil MK, Tanguay PL, Lévesque K, Brégeon J, Mes-Masson AM, Lamarre D, Haibe-Kains B, Trinh VQ, Soucy G, Bilodeau M, Meloche S. Signaling by the tyrosine kinase Yes promotes liver cancer development. Sci Signal 2022; 15:eabj4743. [PMID: 35041461 DOI: 10.1126/scisignal.abj4743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Most patients with hepatocellular carcinoma (HCC) are diagnosed at a late stage and have few therapeutic options and a poor prognosis. This is due to the lack of clearly defined underlying mechanisms or a dominant oncogene that can be targeted pharmacologically, unlike in other cancer types. Here, we report the identification of a previously uncharacterized oncogenic signaling pathway in HCC that is mediated by the tyrosine kinase Yes. Using genetic and pharmacological interventions in cellular and mouse models of HCC, we showed that Yes activity was necessary for HCC cell proliferation. Transgenic expression of activated Yes in mouse hepatocytes was sufficient to induce liver tumorigenesis. Yes phosphorylated the transcriptional coactivators YAP and TAZ (YAP/TAZ), promoting their nuclear accumulation and transcriptional activity in HCC cells and liver tumors. We also showed that YAP/TAZ were effectors of the Yes-dependent oncogenic transformation of hepatocytes. Src family kinase activation correlated with the tyrosine phosphorylation and nuclear localization of YAP in human HCC and was associated with increased tumor burden in mice. Specifically, high Yes activity predicted shorter overall survival in patients with HCC. Thus, our findings identify Yes as a potential therapeutic target in HCC.
Collapse
Affiliation(s)
| | - Marjorie Lapouge
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada.,Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Laure Voisin
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | | | - Pierre-Luc Tanguay
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Kim Lévesque
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Jérémy Brégeon
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Anne-Marie Mes-Masson
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, Quebec, Canada
| | - Daniel Lamarre
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, Quebec, Canada
| | - Benjamin Haibe-Kains
- Departments of Medical Biophysiscs and Computer Science, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Ontario Institute of Cancer Research, Toronto, Ontario, Canada
| | - Vincent Q Trinh
- Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, Quebec, Canada.,Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Geneviève Soucy
- Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, Quebec, Canada.,Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Marc Bilodeau
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Centre hospitalier de l'Université de Montréal (CHUM) Research Centre, Montreal, Quebec, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada.,Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
3
|
Joa H, Blažević T, Grojer C, Zeller I, Heiss EH, Atanasov AG, Feldler I, Gruzdaitis P, Czaloun C, Proksch P, Messner B, Bernhard D, Dirsch VM. Tylophorine reduces protein biosynthesis and rapidly decreases cyclin D1, inhibiting vascular smooth muscle cell proliferation in vitro and in organ culture. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152938. [PMID: 31078367 DOI: 10.1016/j.phymed.2019.152938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 05/13/2023]
Abstract
BACKGROUND Tylophorine (TYL) is an alkaloid with antiproliferative action in cancer cells. Vascular smooth muscle cell (VSMC) proliferation and neointima formation contribute to restenosis after percutaneous coronary interventions. HYPOTHESIS/PURPOSE Our goal was to examine the potential of TYL to inhibit VSMC proliferation and migration, and to dissect underlying signaling pathways. STUDY DESIGN AND METHODS TYL was administered to platelet-derived growth factor (PDGF-BB)-stimulated, serum-stimulated, quiescent and unsynchronized VSMC of rat and human origin. BrdU incorporation and resazurin conversion were used to assess cell proliferation. Cell cycle progression was analyzed by flow cytometry of propidium iodide-stained nuclei. Expression profiles of proteins and mRNAs were determined using western blot analysis and RT-qPCR. The Click-iT OPP Alexa Fluor 488 assay was used to monitor protein biosynthesis. RESULTS TYL inhibited PDGF-BB-induced proliferation of rat aortic VSMCs by arresting cells in G1 phase of the cell cycle with an IC50 of 0.13 µmol/l. The lack of retinoblastoma protein phosphorylation and cyclin D1 downregulation corroborated a G1 arrest. Inhibition of proliferation and cyclin D1 downregulation were species- and stimulus-independent. TYL also decreased levels of p21 and p27 proteins, although at later time points than observed for cyclin D1. Co-treatment of VSMC with TYL and MG132 or cycloheximide (CHX) excluded proteasome activation by TYL as the mechanism of action. Comparable time-dependent downregulation of cyclin D1, p21 and p27 in TYL- or CHX-treated cells, together with decreased protein synthesis observed in the Click-iT assay, suggests that TYL is a protein synthesis inhibitor. Besides proliferation, TYL also suppressed migration of PDGF-activated VSMC. In a human saphenous vein organ culture model for graft disease, TYL potently inhibited intimal hyperplasia. CONCLUSION This unique activity profile renders TYL an interesting lead for the treatment of vasculo-proliferative disorders, such as restenosis.
Collapse
Affiliation(s)
- Helge Joa
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; vasopharm GmbH, Friedrich-Bergius-Ring 15, 97076 Würzburg, Germany
| | - Tina Blažević
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria.
| | - Christoph Grojer
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Pfizer Corp. Austria GmbH, Floridsdorfer Hauptstraße 1, 1210 Wien, Austria
| | - Iris Zeller
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria; Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Elke H Heiss
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria
| | - Atanas G Atanasov
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552, Jastrzebiec, Poland
| | - Ines Feldler
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria
| | - Päivi Gruzdaitis
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Pharmaceutical Information Centre Ltd., Korkeavuorenkatu 35, 00130 Helsinki, Finland
| | - Christa Czaloun
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Auge Gottes Apotheke, Nussdorfer Straße 79, 1090 Wien, Austria
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria
| | - David Bernhard
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria; Center for Medical Research, Johannes Kepler University Linz, Krankenhausstr. 7a, 4020 Linz, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria
| |
Collapse
|
4
|
Chibazakura T, Asano Y. Defective interaction between p27 and cyclin A-CDK complex in certain human cancer cell lines revealed by split YFP assay in living cells. Biosci Biotechnol Biochem 2017; 81:2360-2366. [DOI: 10.1080/09168451.2017.1391686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Abstract
Cyclin–cyclin dependent kinase (CDK) complex is negatively regulated by interaction with CDK inhibitors (CKIs). p27 protein is a major CKI in mammals and its down-regulation correlates with malignant transformation. However, some cancer cells express p27 at normal level, suggesting not only quantitative but qualitative control of p27, although little is known about such control. We analyzed the interaction between p27 and cyclin A (CycA)-CDK complex in living human cell lines, using a split yellow fluorescent protein (YFP) system in which the YFP fluorescence solely depends on p27-CycA binding. Introduction of this system into various cancer cell lines revealed that certain cell lines show no detectable YFP fluorescence. Furthermore, these cell lines exhibited reduced p27-CycA interaction as evaluated by immunoprecipitation, while they showed normal co-localization of both proteins. These results suggest that some cancer cells are defective for efficient interaction between p27 and CycA–CDK complex due to qualitative alteration(s).
Collapse
Affiliation(s)
- Taku Chibazakura
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuichi Asano
- Department of Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
5
|
Yao Y, Hu Z, Ye J, Hu C, Song Q, Da X, Yu Y, Li H, Xu C, Chen Q, Wang QK. Targeting AGGF1 (angiogenic factor with G patch and FHA domains 1) for Blocking Neointimal Formation After Vascular Injury. J Am Heart Assoc 2017. [PMID: 28649088 PMCID: PMC5669188 DOI: 10.1161/jaha.117.005889] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Despite recent improvements in angioplasty and placement of drug‐eluting stents in treatment of atherosclerosis, restenosis and in‐stent thrombosis impede treatment efficacy and cause numerous deaths. Research efforts are needed to identify new molecular targets for blocking restenosis. We aim to establish angiogenic factor AGGF1 (angiogenic factor with G patch and FHA domains 1) as a novel target for blocking neointimal formation and restenosis after vascular injury. Methods and Results AGGF1 shows strong expression in carotid arteries; however, its expression is markedly decreased in arteries after vascular injury. AGGF1+/− mice show increased neointimal formation accompanied with increased proliferation of vascular smooth muscle cells (VSMCs) in carotid arteries after vascular injury. Importantly, AGGF1 protein therapy blocks neointimal formation after vascular injury by inhibiting the proliferation and promoting phenotypic switching of VSMCs to the contractile phenotype in mice in vivo. In vitro, AGGF1 significantly inhibits VSMCs proliferation and decreases the cell numbers at the S phase. AGGF1 also blocks platelet‐derived growth factor‐BB–induced proliferation, migration of VSMCs, increases expression of cyclin D, and decreases expression of p21 and p27. AGGF1 inhibits phenotypic switching of VSMCs to the synthetic phenotype by countering the inhibitory effect of platelet‐derived growth factor‐BB on SRF expression and the formation of the myocardin/SRF/CArG‐box complex involved in activation of VSMCs markers. Finally, we show that AGGF1 inhibits platelet‐derived growth factor‐BB–induced phosphorylation of MEK1/2, ERK1/2, and Elk phosphorylation involved in the phenotypic switching of VSMCs, and that overexpression of Elk abolishes the effect of AGGF1. Conclusions AGGF1 protein therapy is effective in blocking neointimal formation after vascular injury by regulating a novel AGGF1‐MEK1/2‐ERK1/2‐Elk‐myocardin‐SRF/p27 signaling pathway.
Collapse
Affiliation(s)
- Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenkun Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Ye
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Changqing Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qixue Song
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Xingwen Da
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Yubin Yu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyun Chen
- Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH
| | - Qing Kenneth Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China .,Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
6
|
Deubiquitinating Enzyme USP20 Regulates Extracellular Signal-Regulated Kinase 3 Stability and Biological Activity. Mol Cell Biol 2017; 37:MCB.00432-16. [PMID: 28167606 DOI: 10.1128/mcb.00432-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 02/01/2017] [Indexed: 01/17/2023] Open
Abstract
Extracellular signal-regulated kinase 3 (ERK3) is an atypical mitogen-activated protein kinase (MAPK) whose regulatory mechanisms and biological functions remain superficially understood. Contrary to most protein kinases, ERK3 is a highly unstable protein that is subject to dynamic regulation by the ubiquitin-proteasome system. However, the effectors that control ERK3 ubiquitination and degradation are unknown. In this study, we carried out an unbiased functional loss-of-function screen of the human deubiquitinating enzyme (DUB) family and identified ubiquitin-specific protease 20 (USP20) as a novel ERK3 regulator. USP20 interacts with and deubiquitinates ERK3 both in vitro and in intact cells. The overexpression of USP20 results in the stabilization and accumulation of the ERK3 protein, whereas USP20 depletion reduces the levels of ERK3. We found that the expression levels of ERK3 correlate with those of USP20 in various cellular contexts. Importantly, we show that USP20 regulates actin cytoskeleton organization and cell migration in a manner dependent on ERK3 expression. Our results identify USP20 as a bona fide regulator of ERK3 stability and physiological functions.
Collapse
|
7
|
Duhamel S, Girondel C, Dorn JF, Tanguay PL, Voisin L, Smits R, Maddox PS, Meloche S. Deregulated ERK1/2 MAP kinase signaling promotes aneuploidy by a Fbxw7β-Aurora A pathway. Cell Cycle 2016; 15:1631-42. [PMID: 27152455 DOI: 10.1080/15384101.2016.1183851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aneuploidy is a common feature of human solid tumors and is often associated with poor prognosis. There is growing evidence that oncogenic signaling pathways, which are universally dysregulated in cancer, contribute to the promotion of aneuploidy. However, the mechanisms connecting signaling pathways to the execution of mitosis and cytokinesis are not well understood. Here, we show that hyperactivation of the ERK1/2 MAP kinase pathway in epithelial cells impairs cytokinesis, leading to polyploidization and aneuploidy. Mechanistically, deregulated ERK1/2 signaling specifically downregulates expression of the F-box protein Fbxw7β, a substrate-binding subunit of the SCF(Fbxw7) ubiquitin ligase, resulting in the accumulation of the mitotic kinase Aurora A. Reduction of Aurora A levels by RNA interference or pharmacological inhibition of MEK1/2 reverts the defect in cytokinesis and decreases the frequency of abnormal cell divisions induced by oncogenic H-Ras(V12). Reciprocally, overexpression of Aurora A or silencing of Fbxw7β phenocopies the effect of H-Ras(V12) on cell division. In vivo, conditional activation of MEK2 in the mouse intestine lowers Fbxw7β expression, resulting in the accumulation of cells with enlarged nuclei. We propose that the ERK1/2/ Fbxw7β/Aurora A axis identified in this study contributes to genomic instability and tumor progression.
Collapse
Affiliation(s)
- Stéphanie Duhamel
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada.,b Program of Molecular Biology, Université de Montréal , Montreal , Quebec , Canada
| | - Charlotte Girondel
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada.,c Department of Pharmacology , Université de Montréal , Montreal , Quebec , Canada
| | - Jonas F Dorn
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada
| | - Pierre-Luc Tanguay
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada
| | - Laure Voisin
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada
| | - Ron Smits
- d Department of Gastroenterology and Hepatology , Erasmus MC , Rotterdam , The Netherlands
| | - Paul S Maddox
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada.,e Department of Pathology and Cell Biology , Université de Montréal , Montreal , Quebec , Canada
| | - Sylvain Meloche
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montreal , Quebec , Canada.,b Program of Molecular Biology, Université de Montréal , Montreal , Quebec , Canada.,c Department of Pharmacology , Université de Montréal , Montreal , Quebec , Canada
| |
Collapse
|
8
|
Li C, Liu Y, Xie Z, Lu Q, Luo S. Stigmasterol protects against Ang II-induced proliferation of the A7r5 aortic smooth muscle cell-line. Food Funct 2015; 6:2266-72. [DOI: 10.1039/c5fo00031a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Excessive proliferation of vascular smooth muscle cells is a crucial event in the pathogenesis of several cardiovascular diseases, including atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Chunmei Li
- Department of Biochemistry and Molecular Biology
- Guangdong Pharmaceutical College
- Guangzhou
- China
| | - Yang Liu
- Department of Biochemistry and Molecular Biology
- Guangdong Pharmaceutical College
- Guangzhou
- China
| | - Zhe Xie
- Department of Biochemistry and Molecular Biology
- Guangdong Pharmaceutical College
- Guangzhou
- China
| | - Qun Lu
- Department of Biochemistry and Molecular Biology
- Guangdong Pharmaceutical College
- Guangzhou
- China
| | - Shaohong Luo
- Department of Biochemistry and Molecular Biology
- Guangdong Pharmaceutical College
- Guangzhou
- China
| |
Collapse
|
9
|
Andrés V. Vitamin D puts the brakes on angiotensin II-induced oxidative stress and vascular smooth muscle cell senescence. Atherosclerosis 2014; 236:444-7. [PMID: 25173069 DOI: 10.1016/j.atherosclerosis.2014.07.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/26/2014] [Indexed: 02/08/2023]
Abstract
Signaling via both vitamin D (VitD) and the renin-angiotensin system (RAS) plays important roles in physiological processes. Evidence has mounted linking cardiovascular disease to both increased activity of the RAS and VitD deficiency. Although several studies have established functional relationships between the RAS and VitD, many aspects of their complex interaction remain unknown. In this issue of Atherosclerosis, Valcheva and colleagues show that defective VitD signaling can promote vascular damage by inducing premature senescence of smooth muscle cells due to elevated local production of angiotensin II and reactive oxygen species, and upregulation of the tumor suppressor p57(Kip2).
Collapse
Affiliation(s)
- Vicente Andrés
- Laboratory of Molecular and Genetic Cardiovascular Pathophysiology, Department of Atherothrombosis, Imaging and Epidemiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
10
|
Redwan IN, Dyrager C, Solano C, Fernández de Trocóniz G, Voisin L, Bliman D, Meloche S, Grøtli M. Towards the development of chromone-based MEK1/2 modulators. Eur J Med Chem 2014; 85:127-38. [PMID: 25078316 DOI: 10.1016/j.ejmech.2014.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 06/30/2014] [Accepted: 07/06/2014] [Indexed: 11/27/2022]
Abstract
Inhibition or allosteric modulation of mitogen-activated protein kinase kinases MEK1 and MEK2 (MEK1/2) represent a promising strategy for the discovery of new specific anticancer agents. In this paper, structure-based design, beginning from the lead compound PD98059, was used to study potential structural modifications on the chromone structure in order to obtain highly potent derivatives that target the allosteric pocket in MEK1. Subsequently, a small series of PD98059 analogs were synthesized to provide a first generation of chromone-based derivatives that inhibit the activation of MEK1 with IC50 values as low as 30 nM in vitro. Complementary cellular studies also showed that two of the compounds in the series inhibit the activity of MEK1/2 with IC50 values in the nanomolar range (73-97 nM). In addition, compounds in this series were found to inhibit the proliferation of a small panel of human cancer cell lines.
Collapse
Affiliation(s)
- Itedale Namro Redwan
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-41296 Göteborg, Sweden
| | - Christine Dyrager
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-41296 Göteborg, Sweden
| | - Carlos Solano
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-41296 Göteborg, Sweden
| | | | - Laure Voisin
- Institute of Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - David Bliman
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-41296 Göteborg, Sweden
| | - Sylvain Meloche
- Institute of Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Morten Grøtli
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-41296 Göteborg, Sweden.
| |
Collapse
|
11
|
Arts MR, Baron M, Chokr N, Fritzler MJ, the Canadian Scleroderma Research Group (CSRG), Servant MJ. Systemic sclerosis immunoglobulin induces growth and a pro-fibrotic state in vascular smooth muscle cells through the epidermal growth factor receptor. PLoS One 2014; 9:e100035. [PMID: 24927197 PMCID: PMC4057313 DOI: 10.1371/journal.pone.0100035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 05/21/2014] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE It has been suggested that autoantibodies in systemic sclerosis (SSc) may induce the differentiation of cultured fibroblasts into myofibroblasts through platelet-derived growth factor receptor (PDGFR) activation. The present study aims to characterize the effects of SSc IgG on vascular smooth muscle cells (VSMCs) and to determine if stimulatory autoantibodies directed to the PDGFR can be detected, and whether they induce a profibrotic response in primary cultured VSMCs. METHODS Cultured VSMCs were exposed to IgG fractions purified from SSc-patient or control sera. VSMC responses were then analyzed for ERK1/2 and Akt phosphorylation, PDGFR immunoprecipitation, cellular proliferation, protein synthesis, and pro-fibrotic changes in mRNA expression. RESULTS Stimulatory activity in IgG fractions was more prevalent and intense in the SSc samples. SSc IgG immunoprecipitated the PDGFR with greater avidity than control IgG. Interestingly, activation of downstream signaling events (e.g. Akt, ERK1/2) was independent of PDGFR activity, but required functional EGFR. We also detected increased protein synthesis in response to SSc IgG (p<0.001) and pro-fibrotic changes in gene expression (Tgfb1 +200%; Tgfb2 -23%; p<0.001)) in VSMCs treated with SSc IgG. CONCLUSION When compared to control IgG, SSc IgG have a higher stimulation index in VSMCs. Although SSc IgG interact with the PDGFR, the observed remodeling signaling events occur through the EGFR in VSMC. Our data thus favour a model of transactivation of the EGFR by SSc-derived PDGFR autoantibodies and suggest the use of EGFR inhibitors in future target identification studies in the field of SSc.
Collapse
MESH Headings
- Adult
- Animals
- Cell Proliferation/drug effects
- Cells, Cultured
- ErbB Receptors/physiology
- Female
- Fibrosis/chemically induced
- Humans
- Immunoglobulin G/pharmacology
- Male
- Middle Aged
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Rats
- Rats, Wistar
- Scleroderma, Systemic/immunology
- Scleroderma, Systemic/pathology
Collapse
Affiliation(s)
- Monique R. Arts
- Université de Montréal, Faculté de Pharmacie, Montreal, Quebec, Canada
- McGill University, Jewish General Hospital, Montreal, Quebec, Canada
| | - Murray Baron
- McGill University, Jewish General Hospital, Montreal, Quebec, Canada
| | - Nidaa Chokr
- Université de Montréal, Faculté de Pharmacie, Montreal, Quebec, Canada
| | | | | | - Marc J. Servant
- Université de Montréal, Faculté de Pharmacie, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Marquis M, Boulet S, Mathien S, Rousseau J, Thébault P, Daudelin JF, Rooney J, Turgeon B, Beauchamp C, Meloche S, Labrecque N. The non-classical MAP kinase ERK3 controls T cell activation. PLoS One 2014; 9:e86681. [PMID: 24475167 PMCID: PMC3903551 DOI: 10.1371/journal.pone.0086681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 12/13/2013] [Indexed: 11/19/2022] Open
Abstract
The classical mitogen-activated protein kinases (MAPKs) ERK1 and ERK2 are activated upon stimulation of cells with a broad range of extracellular signals (including antigens) allowing cellular responses to occur. ERK3 is an atypical member of the MAPK family with highest homology to ERK1/2. Therefore, we evaluated the role of ERK3 in mature T cell response. Mouse resting T cells do not transcribe ERK3 but its expression is induced in both CD4⁺ and CD8⁺ T cells following T cell receptor (TCR)-induced T cell activation. This induction of ERK3 expression in T lymphocytes requires activation of the classical MAPK ERK1 and ERK2. Moreover, ERK3 protein is phosphorylated and associates with MK5 in activated primary T cells. We show that ERK3-deficient T cells have a decreased proliferation rate and are impaired in cytokine secretion following in vitro stimulation with low dose of anti-CD3 antibodies. Our findings identify the atypical MAPK ERK3 as a new and important regulator of TCR-induced T cell activation.
Collapse
Affiliation(s)
- Miriam Marquis
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Quebec, Canada
| | - Salix Boulet
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - Simon Mathien
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | - Justine Rousseau
- Department of Pharmacology and Molecular Biology, University of Montreal, Quebec, Canada
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | - Paméla Thébault
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | | | - Julie Rooney
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - Benjamin Turgeon
- Department of Pharmacology and Molecular Biology, University of Montreal, Quebec, Canada
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | | | - Sylvain Meloche
- Department of Pharmacology and Molecular Biology, University of Montreal, Quebec, Canada
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Quebec, Canada
| |
Collapse
|
13
|
Lee M, Pellegata NS. Multiple endocrine neoplasia syndromes associated with mutation of p27. J Endocrinol Invest 2013; 36:781-7. [PMID: 23800691 DOI: 10.3275/9021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Multiple endocrine neoplasias (MEN) are autosomal dominant disorders characterized by the occurrence of tumors in at least two endocrine glands. Until recently, two MEN syndromes were known, i.e. the MEN type 1 (MEN1) and type 2 (MEN2), which are caused by germline mutations in the MEN1 and RET genes, respectively. These two syndromes are characterized by a different tumor spectrum. A few years ago we described a variant of the MEN syndromes, which spontaneously developed in a rat colony and was named MENX. Affected animals consistently develop multiple endocrine tumors, with a spectrum that shares features with both MEN1 and MEN2 human syndromes. Genetic studies identified a germline mutation in the Cdkn1b gene, encoding the p27 cell cycle inhibitor, as the causative mutation for MENX. Capitalizing on these findings, germline mutations in the human homologue, CDKN1B, were searched for and identified in patients with multiple endocrine tumors. As a consequence of this discovery, a novel human MEN syndrome, named MEN4, was recognized, which is caused by heterozygous mutations in p27. These studies identified Cdkn1b/CDKN1B as a novel tumor susceptibility gene for multiple endocrine tumors in both rats and humans. Here we review the characteristics of the MENX and MEN4 syndromes and we briefly address the main function of p27 and how it is affected by MENX- or MEN4-associated mutations.
Collapse
Affiliation(s)
- M Lee
- Institute of Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | | |
Collapse
|
14
|
Phosphoproteome dynamics reveal novel ERK1/2 MAP kinase substrates with broad spectrum of functions. Mol Syst Biol 2013; 9:669. [PMID: 23712012 PMCID: PMC4188273 DOI: 10.1038/msb.2013.25] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 04/18/2013] [Indexed: 01/23/2023] Open
Abstract
The ERK1/2 MAP kinase pathway is an evolutionarily conserved signaling module that controls many fundamental physiological processes. Deregulated activity of ERK1/2 MAP kinases is associated with developmental syndromes and several human diseases. Despite the importance of this pathway, a comprehensive picture of the natural substrate repertoire and biochemical mechanisms regulated by ERK1/2 is still lacking. In this study, we used large-scale quantitative phosphoproteomics and bioinformatics analyses to identify novel candidate ERK1/2 substrates based on their phosphorylation signature and kinetic profiles in epithelial cells. We identified a total of 7936 phosphorylation sites within 1861 proteins, of which 155 classify as candidate ERK1/2 substrates, including 128 new targets. Candidate ERK1/2 substrates are involved in diverse cellular processes including transcriptional regulation, chromatin remodeling, RNA splicing, cytoskeleton dynamics, cellular junctions and cell signaling. Detailed characterization of one newly identified substrate, the transcriptional regulator JunB, revealed that ERK1/2 phosphorylate JunB on a serine adjacent to the DNA-binding domain, resulting in increased DNA-binding affinity and transcriptional activity. Our study expands the spectrum of cellular functions controlled by ERK1/2 kinases.
Collapse
|
15
|
Handy I, Patel RC. STAT1 requirement for PKR-induced cell cycle arrest in vascular smooth muscle cells in response to heparin. Gene 2013; 524:15-21. [PMID: 23597922 DOI: 10.1016/j.gene.2013.03.124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 03/18/2013] [Accepted: 03/27/2013] [Indexed: 11/27/2022]
Abstract
Interferons (IFNs) are a family of cytokines that exhibit antiviral, antiproliferative, and immunomodulatory properties. PKR (protein kinase, RNA activated) is of central importance in mediating the antiproliferative actions of IFNs. Our research has established that PKR inhibits vascular smooth muscle cell (VSMC) proliferation by regulating G1 to S transition. Many cardiovascular diseases result from complications of atherosclerosis, a chronic and progressive inflammatory condition often characterized by excessive proliferation of VSMC. Thus, an effective method for inhibiting VSMC proliferation is likely to arrest atherosclerosis and restenosis at early stages. Our research establishes that PKR activation in VSMC leads to a G1 arrest brought about by an inhibition of cyclin-dependent kinase 2 (Cdk2) activity by p27(kip1). In quiescent VSMC, p27(kip1) levels are high and when stimulated by serum/growth factors, p27(kip1) levels drop by destabilization of the protein. Under conditions that lead to activation of PKR, there is a marked inhibition of p27(kip1) down-regulation due to increased stability of p27(kip1) protein. In order to understand the mechanism of heparin-induced stabilization of p27(kip1) in VSMC, we examined the involvement of the Signal Transducer and Activator of Transcription-1 (STAT1), which is an important player in mediating antiproliferative effects of IFNs. Our results demonstrate that PKR overexpression in VSMC leads to an increase in p27(kip1) protein levels and this increase requires the catalytic activity of PKR. PKR activation induced by antiproliferative agent heparin leads to phosphorylation of STAT1 on serine 727, which is essential for the cell cycle block. STAT1 null VSMCs are largely defective in heparin-induced cell cycle arrest and in PKR null cells the STAT1 phosphorylation in response to heparin was absent. These results establish that heparin causes STAT1 phosphorylation on serine 727 via activation of PKR and that this event is required for the G1 arrest in VSMC.
Collapse
Affiliation(s)
- Indhira Handy
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208,USA
| | | |
Collapse
|
16
|
Pandhare J, Mantri C, Gong Y, Chen Z, Dash C. XMRV accelerates cellular proliferation, transformational activity, and invasiveness of prostate cancer cells by downregulating p27(Kip1). Prostate 2012; 72:886-97. [PMID: 21932423 PMCID: PMC3275676 DOI: 10.1002/pros.21491] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 08/24/2011] [Indexed: 12/29/2022]
Abstract
BACKGROUND Xenotropic murine leukemia virus-related retrovirus (XMRV) is a recently discovered gammaretrovirus that was originally detected in prostate tumors. However, a causal relationship between XMRV and prostate cancer remains controversial due to conflicting reports on its etiologic occurrence. Even though gammaretroviruses are known to induce cancer in animals, a mechanism for XMRV-induced carcinogenesis remains unknown. Several mechanisms including insertional mutagenesis, proinflammatory effects, oncogenic viral proteins, immune suppression, and altered epithelial/stromal interactions have been proposed for a role of XMRV in prostate cancer. However, biochemical data supporting any of these mechanisms are lacking. Therefore, our aim was to evaluate a potential role of XMRV in prostate carcinogenesis. METHODS Growth kinetics of prostate cancer cells are conducted by MTT assay. In vitro transformation and invasion was carried out by soft agar colony formation, and Matrigel cell invasion assay, respectively. p27(Kip1) expression was determined by Western blot and MMP activation was evaluated by gelatin-zymography. Up-regulation of miR221 and miR222 expression was examined by real-time PCR. RESULTS We demonstrate that XMRV infection can accelerate cellular proliferation, enhance transformation, and increase invasiveness of slow growing prostate cancer cells. The molecular basis of these viral induced activities is mediated by the downregulation of cyclin/cyclin dependent kinase inhibitor p27(Kip1) . Downstream analyses illustrated that XMRV infection upregulates miR221 and miR222 expression that target p27(Kip1) mRNA. CONCLUSIONS We propose that downregulation of p27(Kip1) by XMRV infection facilitates transition of G1 to S, thereby accelerates growth of prostate cancer cells. Our findings implicate that if XMRV is present in humans, then under appropriate cellular microenvironment it may serve as a cofactor to promote cancer progression in the prostate.
Collapse
Affiliation(s)
- Jui Pandhare
- Laboratory of Retrovirology and Epigenetics, Center for AIDS Health Disparities Research, Vanderbilt-Meharry Center For AIDS Research (CFAR), Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
| | - Chinmay Mantri
- Laboratory of Retrovirology and Epigenetics, Center for AIDS Health Disparities Research, Vanderbilt-Meharry Center For AIDS Research (CFAR), Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
| | - Yuanying Gong
- Department of Biochemistry and Cancer Biology, Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
| | - Chandravanu Dash
- Laboratory of Retrovirology and Epigenetics, Center for AIDS Health Disparities Research, Vanderbilt-Meharry Center For AIDS Research (CFAR), Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College School of Medicine, 1005 Dr. DB Todd Jr Blvd., Nashville, TN 37208, USA
- Corresponding Author: Tel: 615-327-6996, Fax: 615-327-6929,
| |
Collapse
|
17
|
Abstract
Multiple endocrine neoplasias are autosomal dominant disorders characterized by the occurrence of tumors in at least two endocrine glands. Two MEN syndromes have long been known and are well characterized: the MEN type 1 (MEN1) and type 2 (MEN2). These syndromes are caused by germline mutations in the MEN1 and RET genes, respectively, and have a different tumor spectrum. Recently, a variant of the MEN syndromes arose spontaneously in a rat colony and was named MENX. Affected animals consistently develop multiple endocrine tumors, with a spectrum that shares features with both MEN1 and MEN2 human syndromes. Genetic studies identified a germline mutation in the Cdkn1b gene, encoding the p27 cell cycle inhibitor, as the causative mutation for MENX. Capitalizing on these findings, heterozygous germline mutations in the human homologue, CDKN1B, were searched for and identified in patients with multiple endocrine tumors. As a consequence of this discovery, a novel human MEN syndrome, named MEN4, was recognized, which is caused by mutations in p27. Altogether, these studies identified Cdkn1b/CDKN1B as a novel tumor susceptibility gene for multiple endocrine tumors in both rats and humans. Here we review the characteristics of the MENX and MEN4 syndromes and we briefly address the main function of p27 and how they are affected by MENX/4-associated mutations.
Collapse
Affiliation(s)
- Natalia S Pellegata
- Institute of Pathology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
18
|
Fang Z, Zhang T, Dizeyi N, Chen S, Wang H, Swanson KD, Cai C, Balk SP, Yuan X. Androgen Receptor Enhances p27 Degradation in Prostate Cancer Cells through Rapid and Selective TORC2 Activation. J Biol Chem 2011; 287:2090-8. [PMID: 22139837 DOI: 10.1074/jbc.m111.323303] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Androgen receptor (AR) plays a central role in prostate cancer (PCa) growth, with androgen deprivation or AR down-regulation causing cell-cycle arrest and accumulation of the p27 cyclin-dependent kinase inhibitor. The molecular basis for this AR regulation of cell-cycle progression remains unclear. Here we demonstrate that androgen can rapidly reduce p27 protein in PCa cells by increasing its proteasome-mediated degradation. This rapid androgen-stimulated p27 degradation was mediated by AKT through the phosphorylation of p27 T157. Significantly, androgen increased TORC2-mediated AKT S473 phosphorylation without affecting the PDK1-mediated AKT T308 phosphorylation or TORC1 activity. The TORC2 activation was further supported by enhanced mTOR/RICTOR association and increased phosphorylation of additional TORC2 substrates, SGK1 and PKCα. The androgen-stimulated nuclear translocation of AR was associated with markedly-increased nuclear SIN1, a critical component of TORC2. Finally, the androgen-mediated TORC2/AKT activation targets a subset of AKT substrates including p27 and FOXO1, but not PRAS40. This study reveals a pathway linking AR to a selective activation of TORC2, the subsequent activation of AKT, and phosphorylation of a discrete set of AKT substrates that regulate cellular proliferation and survival. These findings establish that TORC2 can function as a central regulator of growth in response to signals that are distinct from those regulating TORC1, and support efforts to target TORC2 for cancer therapy.
Collapse
Affiliation(s)
- Zi Fang
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Platonin inhibited PDGF-BB-induced proliferation of rat vascular smooth muscle cells via JNK1/2-dependent signaling. Acta Pharmacol Sin 2011; 32:1337-44. [PMID: 21892199 DOI: 10.1038/aps.2011.105] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
AIM To examine the inhibitory actions of the immunoregulator platonin against proliferation of rat vascular smooth muscle cells (VSMCs). METHODS VSMCs were prepared from the thoracic aortas of male Wistar rats. Cell proliferation was examined using MTT assays. Cell cycles were analyzed using flow cytometry. c-Jun N-terminal kinase (JNK)1/2, extracellular signal-regulated kinase (ERK)1/2, AKT, and c-Jun phosphorylation or p27 expression were detected using immunoblotting. RESULTS Pretreatment with platonin (1-5 μmol/L) significantly suppressed VSMC proliferation stimulated by PDGF-BB (10 ng/mL) or 10% fetal bovine serum (FBS), and arrested cell cycle progression in the S and G(2)/M phases. The same concentrations of platonin significantly inhibited the phosphorylation of JNK1/2 but not ERK1/2 or AKT in VSMCs stimulated by PDGF-BB. Furthermore, platonin also attenuated c-Jun phosphorylation and markedly reversed the down-regulation of p27 expression after PDGF-BB stimulation. CONCLUSION Platonin inhibited VSMC proliferation, possibly via inhibiting phosphorylation of JNK1/2 and c-Jun, and reversal of p27 down-regulation, thereby leading to cell cycle arrest at the S and G(2)/M phases. Thus, platonin may represent a novel approach for lowering the risk of abnormal VSMC proliferation and related vascular diseases.
Collapse
|
20
|
An allosteric inhibitor of the human Cdc34 ubiquitin-conjugating enzyme. Cell 2011; 145:1075-87. [PMID: 21683433 DOI: 10.1016/j.cell.2011.05.039] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 05/02/2011] [Accepted: 05/31/2011] [Indexed: 12/29/2022]
Abstract
In the ubiquitin-proteasome system (UPS), E2 enzymes mediate the conjugation of ubiquitin to substrates and thereby control protein stability and interactions. The E2 enzyme hCdc34 catalyzes the ubiquitination of hundreds of proteins in conjunction with the cullin-RING (CRL) superfamily of E3 enzymes. We identified a small molecule termed CC0651 that selectively inhibits hCdc34. Structure determination revealed that CC0651 inserts into a cryptic binding pocket on hCdc34 distant from the catalytic site, causing subtle but wholesale displacement of E2 secondary structural elements. CC0651 analogs inhibited proliferation of human cancer cell lines and caused accumulation of the SCF(Skp2) substrate p27(Kip1). CC0651 does not affect hCdc34 interactions with E1 or E3 enzymes or the formation of the ubiquitin thioester but instead interferes with the discharge of ubiquitin to acceptor lysine residues. E2 enzymes are thus susceptible to noncatalytic site inhibition and may represent a viable class of drug target in the UPS.
Collapse
|
21
|
Tudev M, Lim Y, Park ES, Kim WS, Lim IH, Kwak JH, Jung JK, Hong JT, Yoo HS, Lee MY, Pyo MY, Yun YP. Anti-Proliferative Activity of OD78 Is Mediated through Cell Cycle Progression by Upregulation p27 kip1in Rat Aortic Vascular Smooth Muscle Cells. Biomol Ther (Seoul) 2011. [DOI: 10.4062/biomolther.2011.19.2.187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
22
|
Perez J, Torres RA, Rocic P, Cismowski MJ, Weber DS, Darley-Usmar VM, Lucchesi PA. PYK2 signaling is required for PDGF-dependent vascular smooth muscle cell proliferation. Am J Physiol Cell Physiol 2011; 301:C242-51. [PMID: 21451101 DOI: 10.1152/ajpcell.00315.2010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Aberrant vascular smooth muscle cell (VSMC) growth is associated with many vascular diseases including atherosclerosis, hypertension, and restenosis. Platelet-derived growth factor-BB (PDGF) induces VSMC proliferation through control of cell cycle progression and protein and DNA synthesis. Multiple signaling cascades control VSMC growth, including members of the mitogen-activated protein kinase (MAPK) family as well as phosphatidylinositol 3-kinase (PI3K) and its downstream effector AKT/protein kinase B (PKB). Little is known about how these signals are integrated by mitogens and whether there are common receptor-proximal signaling control points that synchronize the execution of physiological growth functions. The nonreceptor proline-rich tyrosine kinase 2 (PYK2) is activated by a variety of growth factors and G protein receptor agonists in VSMC and lies upstream of both PI3K and MAPK cascades. The present study investigated the role of PYK2 in PDGF signaling in cultured rat aortic VSMC. PYK2 downregulation attenuated PDGF-dependent protein and DNA synthesis, which correlated with inhibition of AKT and extracellular signal-regulated kinases 1 and 2 (ERK1/2) but not p38 MAPK activation. Inhibition of PDGF-dependent protein kinase B (AKT) and ERK1/2 signaling by inhibitors of upstream kinases PI3K and MEK, respectively, as well as downregulation of PYK2 resulted in modulation of the G(1)/S phase of the cell cycle through inhibition of retinoblastoma protein (Rb) phosphorylation and cyclin D(1) expression, as well as p27(Kip) upregulation. Cell division kinase 2 (cdc2) phosphorylation at G(2)/M was also contingent on PDGF-dependent PI3K-AKT and ERK1/2 signaling. These data suggest that PYK2 is an important upstream mediator in PDGF-dependent signaling cascades that regulate VSMC proliferation.
Collapse
Affiliation(s)
- Jessica Perez
- Department of Cell Biology, University of Alabama at Birmingham, Alabama, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Marinoni I, Pellegata NS. p27kip1: a new multiple endocrine neoplasia gene? Neuroendocrinology 2011; 93:19-28. [PMID: 20980721 DOI: 10.1159/000320366] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 08/18/2010] [Indexed: 11/19/2022]
Abstract
Multiple endocrine neoplasias (MEN) are autosomal dominant disorders characterized by the occurrence of tumors in at least two endocrine glands. Two types of MEN syndromes have long been known: MEN type 1 (MEN1) and MEN type 2 (MEN2), associated with a different spectrum of affected organs. MEN1 and MEN2 are caused by germline mutations in the MEN1 tumor suppressor gene and the RET proto-oncogene, respectively. Lately, a new type of MEN was identified (named MEN4) which is due to mutations in the CDKN1B gene, encoding for p27kip1 (p27), a cyclin-dependent kinase (Cdk) inhibitor that regulates the transition of cells from G1 to S phase. p27 is a non-canonical tumor suppressor since it is usually not somatically mutated in human cancers but it is often downregulated by post-translational mechanisms. The discovery of MEN4 has defined a new role for CDKN1B as a tumor susceptibility gene for multiple endocrine tumors. To date, six germline CDKN1B mutations have been found in patients with a MEN1-like phenotype but negative for MEN1 mutations. Due to the limited number of patients so far identified, the phenotypic features of MEN4 are not clearly defined. Here, we review the clinical and molecular characteristics of the MEN4 syndrome and summarize the main functions of p27 to better comprehend how their alteration can predispose to neuroendocrine tumors.
Collapse
Affiliation(s)
- Ilaria Marinoni
- Institute of Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | | |
Collapse
|
24
|
Déléris P, Trost M, Topisirovic I, Tanguay PL, Borden KLB, Thibault P, Meloche S. Activation loop phosphorylation of ERK3/ERK4 by group I p21-activated kinases (PAKs) defines a novel PAK-ERK3/4-MAPK-activated protein kinase 5 signaling pathway. J Biol Chem 2010; 286:6470-8. [PMID: 21177870 DOI: 10.1074/jbc.m110.181529] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Classical mitogen-activated protein (MAP) kinases are activated by dual phosphorylation of the Thr-Xxx-Tyr motif in their activation loop, which is catalyzed by members of the MAP kinase kinase family. The atypical MAP kinases extracellular signal-regulated kinase 3 (ERK3) and ERK4 contain a single phospho-acceptor site in this segment and are not substrates of MAP kinase kinases. Previous studies have shown that ERK3 and ERK4 are phosphorylated on activation loop residue Ser-189/Ser-186, resulting in their catalytic activation. However, the identity of the protein kinase mediating this regulatory event has remained elusive. We have used an unbiased biochemical purification approach to isolate the kinase activity responsible for ERK3 Ser-189 phosphorylation. Here, we report the identification of group I p21-activated kinases (PAKs) as ERK3/ERK4 activation loop kinases. We show that group I PAKs phosphorylate ERK3 and ERK4 on Ser-189 and Ser-186, respectively, both in vitro and in vivo, and that expression of activated Rac1 augments this response. Reciprocally, silencing of PAK1/2/3 expression by RNA interference (RNAi) completely abolishes Rac1-induced Ser-189 phosphorylation of ERK3. Importantly, we demonstrate that PAK-mediated phosphorylation of ERK3/ERK4 results in their enzymatic activation and in downstream activation of MAP kinase-activated protein kinase 5 (MK5) in vivo. Our results reveal that group I PAKs act as upstream activators of ERK3 and ERK4 and unravel a novel PAK-ERK3/ERK4-MK5 signaling pathway.
Collapse
Affiliation(s)
- Paul Déléris
- Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Targeted inactivation of Mapk4 in mice reveals specific nonredundant functions of Erk3/Erk4 subfamily mitogen-activated protein kinases. Mol Cell Biol 2010; 30:5752-63. [PMID: 20956558 DOI: 10.1128/mcb.01147-10] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Erk4 and Erk3 are atypical members of the mitogen-activated protein (MAP) kinase family. The high sequence identity of Erk4 and Erk3 proteins and the similar organization of their genes imply that the two protein kinases are paralogs. Recently, we have shown that Erk3 function is essential for neonatal survival and critical for the establishment of fetal growth potential and pulmonary function. To investigate the specific functions of Erk4, we have generated mice with a targeted disruption of the Mapk4 gene. We show that Erk4-deficient mice are viable and fertile and exhibit no gross morphological or physiological anomalies. Loss of Erk4 is not compensated by changes in Erk3 expression or activity during embryogenesis or in adult tissues. We further demonstrate that additional loss of Erk4 does not exacerbate the fetal growth restriction and pulmonary immaturity phenotypes of Erk3(-/-) mice and does not compromise the viability of Erk3(+/-) neonates. Interestingly, behavioral phenotyping revealed that Erk4-deficient mice manifest depression-like behavior in the forced-swimming test. Our analysis indicates that the MAP kinase Erk4 is dispensable for mouse embryonic development and reveals that Erk3 and Erk4 have acquired specialized functions through evolutionary diversification.
Collapse
|
26
|
Doyon P, Servant MJ. Tumor necrosis factor receptor-associated factor-6 and ribosomal S6 kinase intracellular pathways link the angiotensin II AT1 receptor to the phosphorylation and activation of the IkappaB kinase complex in vascular smooth muscle cells. J Biol Chem 2010; 285:30708-18. [PMID: 20659889 PMCID: PMC2945565 DOI: 10.1074/jbc.m110.126433] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of NF-κB transcription factors by locally produced angiotensin II (Ang II) is proposed to be involved in chronic inflammatory reactions leading to atherosclerosis development. However, a clear understanding of the signaling cascades coupling the Ang II AT1 receptors to the activation of NF-κB transcription factors is still lacking. Using primary cultured aortic vascular smooth muscle cells, we show that activation of the IKK complex and NF-κB transcription factors by Ang II is regulated by phosphorylation of the catalytic subunit IKKβ on serine residues 177 and 181 in the activation T-loop. The use of pharmacological inhibitors against conventional protein kinases C (PKCs), mitogen-activated/extracellular signal-regulated kinase (MEK) 1/2, ribosomal S6 kinase (RSK), and silencing RNA technology targeting PKCα, IKKβ subunit, tumor growth factor β-activating kinase-1 (TAK1), the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor-6 (TRAF6), and RSK isoforms, demonstrates the requirement of two distinct signaling pathway for the phosphorylation of IKKβ and the activation of the IKK complex by Ang II. Rapid phosphorylation of IKKβ requires a second messenger-dependent pathway composed of PKCα-TRAF6-TAK1, whereas sustained phosphorylation and activation of IKKβ requires the MEK1/2-ERK1/2-RSK pathway. Importantly, simultaneously targeting components of these two pathways completely blunts the phosphorylation of IKKβ and the proinflammatory effect of the octapeptide. This is the first report demonstrating activation of TAK1 by the AT1R. We propose a model whereby TRAF6-TAK1 and ERK-RSK intracellular pathways independently and sequentially converge to the T-loop phosphorylation for full activation of IKKβ, which is an essential step in the proinflammatory activity of Ang II.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/pathology
- Cell Line
- Enzyme Activation/drug effects
- Humans
- I-kappa B Kinase/metabolism
- Inflammation/metabolism
- Inflammation/pathology
- MAP Kinase Kinase 1/antagonists & inhibitors
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/antagonists & inhibitors
- MAP Kinase Kinase 2/metabolism
- MAP Kinase Kinase Kinases/antagonists & inhibitors
- MAP Kinase Kinase Kinases/metabolism
- MAP Kinase Signaling System
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/metabolism
- Models, Biological
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phosphorylation/drug effects
- Protein Kinase C-alpha/antagonists & inhibitors
- Protein Kinase C-alpha/metabolism
- Protein Kinase Inhibitors/pharmacology
- Rats
- Receptor, Angiotensin, Type 1/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Second Messenger Systems
- TNF Receptor-Associated Factor 6/metabolism
Collapse
Affiliation(s)
- Priscilla Doyon
- From the Faculty of Pharmacy and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montreal H3C 3J7, Canada
| | - Marc J. Servant
- From the Faculty of Pharmacy and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montreal H3C 3J7, Canada
- Holds a Canada Research Chair in signalling networks in inflammation. To whom correspondence should be addressed: C.P. 6128, succursale Centre-Ville, Montréal, Québec H3C 3J7, Canada. Tel.: 514-343-7966; Fax: 514-343-7073; E-mail:
| |
Collapse
|
27
|
Han SJ, Lee JH, Choi KY, Hong SH. Novel p104 protein regulates cell proliferation through PI3K inhibition and p27(Kip1) expression. BMB Rep 2010; 43:199-204. [PMID: 20356461 DOI: 10.5483/bmbrep.2010.43.3.199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The protein p104 was first isolated as a binding partner of the Src homology domain of phospholipase Cgamma1, and has been shown to associate with p85alpha, Grb2. The ectopic expression of p104 reduced cellular growth rate, which was also achieved with the overexpression of only the proline-rich region of p104. The proline-rich region of p104 has been found to inhibit the colony formation of platelet-derived growth factor BB-stimulated NIH3T3 cells and MCF7 cancer cells on soft agar. Mutagenesis analysis showed that the second and third proline-rich regions are essential for growth control, as well as for interaction with p85alpha. Overexpression of p104 increased the level of the cyclin-dependent kinase inhibitor, p27(Kip1), and inhibited the activity of phosphoinositide 3-kinase (PI3K). In summary, p104 interacts with p85alpha and is involved in the regulation of p27(Kip1) expression for the reduction of cellular proliferation.
Collapse
Affiliation(s)
- Seung Jin Han
- Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | | | | | | |
Collapse
|
28
|
C-terminal domain phosphorylation of ERK3 controlled by Cdk1 and Cdc14 regulates its stability in mitosis. Biochem J 2010; 428:103-11. [PMID: 20236090 DOI: 10.1042/bj20091604] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
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.
Collapse
|
29
|
Genetic demonstration of a redundant role of extracellular signal-regulated kinase 1 (ERK1) and ERK2 mitogen-activated protein kinases in promoting fibroblast proliferation. Mol Cell Biol 2010; 30:2918-32. [PMID: 20368360 DOI: 10.1128/mcb.00131-10] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The extracellular signal-regulated kinase 1 and 2 (ERK1/2) mitogen-activated protein (MAP) kinase signaling pathway plays an important role in the proliferative response of mammalian cells to mitogens. However, the individual contribution of the isoforms ERK1 and ERK2 to cell proliferation control is unclear. The two ERK isoforms have similar biochemical properties and recognize the same primary sequence determinants on substrates. On the other hand, analysis of mice lacking individual ERK genes suggests that ERK1 and ERK2 may have evolved unique functions. In this study, we used a robust genetic approach to analyze the individual functions of ERK1 and ERK2 in cell proliferation using genetically matched primary embryonic fibroblasts. We show that individual loss of either ERK1 or ERK2 slows down the proliferation rate of fibroblasts to an extent reflecting the expression level of the kinase. Moreover, RNA interference-mediated silencing of ERK1 or ERK2 expression in cells genetically disrupted for the other isoform similarly reduces cell proliferation. We generated fibroblasts genetically deficient in both Erk1 and Erk2. Combined loss of ERK1 and ERK2 resulted in a complete arrest of cell proliferation associated with G(1) arrest and premature replicative senescence. Together, our findings provide compelling genetic evidence for a redundant role of ERK1 and ERK2 in promoting cell proliferation.
Collapse
|
30
|
Khattar E, Kumar V. Mitogenic regulation of p27(Kip1) gene is mediated by AP-1 transcription factors. J Biol Chem 2009; 285:4554-61. [PMID: 19959471 DOI: 10.1074/jbc.m109.029280] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The abundance of cyclin-dependent kinase inhibitor p27(Kip1) during the cell cycle determines whether cells will proliferate or become quiescent. Although the post-translational regulation of p27(Kip1) is well established, its transcriptional regulation is poorly understood. Here, we report that mitogenic stimulation of quiescent HEK293 and Huh7 cells showed a rapid decline in the levels of p27(Kip1) transcript by 2.4 +/- 0.1-fold. Inhibition of the p27(Kip1) gene in response to mitogens involved transcriptional down-regulation and required newly synthesized protein(s). Mutation of the AP-1 element at position -469 in the human p27(Kip1) promoter abrogated the effect of mitogens. The recruitment of the AP-1 complex to the p27(Kip1) promoter was confirmed by in vitro DNA binding and chromatin immunoprecipitation studies. Reporter gene analysis combined with enforced expression of Jun/Fos proteins suggested the involvement of Jun/Fos heterodimer in the transrepression process. Both MAPK and phosphatidylinositol 3-kinase signaling pathways appeared to mediate p27(Kip1) transcription. Furthermore, hepatitis B virus X protein-mediated down-regulation of p27(Kip1) in a transgenic environment correlated with an increase in c-Fos levels, reiterating the physiological relevance of AP-1 in the transcriptional regulation of p27(Kip1). Collectively, our studies present the first evidence demonstrating the role of the AP-1 complex in transcriptional down-regulation of the p27(Kip1) gene following mitogenic stimulation.
Collapse
Affiliation(s)
- Ekta Khattar
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | |
Collapse
|
31
|
Panda D, Goltzman D, Jüppner H, Karaplis AC. TIP39/parathyroid hormone type 2 receptor signaling is a potent inhibitor of chondrocyte proliferation and differentiation. Am J Physiol Endocrinol Metab 2009; 297:E1125-36. [PMID: 19706789 PMCID: PMC2781349 DOI: 10.1152/ajpendo.00254.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Tuberoinfundibular peptide of 39 residues (TIP39) is a member of the parathyroid hormone (PTH) family of peptide hormones that exerts its function by interacting with the PTH type 2 receptor (PTH2R). Presently, no known function has been attributed to this signaling pathway in the developing skeleton. We observed that TIP39 and PTH2R were present in the newborn mouse growth plate, with the receptor localizing in the resting zone whereas ligand expression was restricted exclusively in prehypertrophic and hypertrophic chondrocytes. By 8 wk of life, PTH2R, and to a lesser degree TIP39, immunoreactivity was present in articular chondrocytes. We therefore sought to investigate the role of TIP39/PTH2R signaling in chondrocytes by generating stably transfected CFK2 chondrocytic cells overexpressing PTH2R (CFK2R). TIP39 treatment of CFK2R clones in culture inhibited their proliferation by restricting cells at the G(0)/G(1) phase of the cell cycle, coupled with decreased expression and activity of cyclin-dependent kinases Cdk2 and Cdk4, while p21, an inhibitor of Cdks, was upregulated. In addition, TIP39 treatment decreased expression of differentiation markers in these cells associated with marked alterations in extracellular matrix and metalloproteinase expression. Transcription of Sox9, the master regulator of cartilage differentiation, was reduced in TIP39-treated CFK2R clones. Moreover, Sox9 promoter activity, as measured by luciferase reporter assay, was markedly diminished after TIP39 treatment. In summary, our results show that TIP39/PTH2R signaling inhibits proliferation and alters differentiation of chondrocytes by modulating SOX9 expression, thereby substantiating the functional significance of this signaling pathway in chondrocyte biology.
Collapse
Affiliation(s)
- Dibiyendu Panda
- Div. of Endocrinology, Dept. of Medicine, Sir Mortimer B. Davis-Jewish General Hosp., 3755 Cote Ste Catherine Road, Montreal, QC, Canada H3T 1E2
| | | | | | | |
Collapse
|
32
|
Granados DP, Tanguay PL, Hardy MP, Caron E, de Verteuil D, Meloche S, Perreault C. ER stress affects processing of MHC class I-associated peptides. BMC Immunol 2009; 10:10. [PMID: 19220912 PMCID: PMC2657905 DOI: 10.1186/1471-2172-10-10] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 02/16/2009] [Indexed: 12/19/2022] Open
Abstract
Background Viral infection and neoplastic transformation trigger endoplasmic reticulum (ER) stress. Thus, a large proportion of the cells that must be recognized by the immune system are stressed cells. Cells respond to ER stress by launching the unfolded protein response (UPR). The UPR regulates the two key processes that control major histocompatibility complex class I (MHC I)-peptide presentation: protein synthesis and degradation. We therefore asked whether and how the UPR impinges on MHC I-peptide presentation. Results We evaluated the impact of the UPR on global MHC I expression and on presentation of the H2Kb-associated SIINFEKL peptide. EL4 cells stably transfected with vectors coding hen egg lysozyme (HEL)-SIINFEKL protein variants were stressed with palmitate or exposed to glucose deprivation. UPR decreased surface expression of MHC I but did not affect MHC I mRNA level nor the total amount of intracellular MHC I proteins. Impaired MHC I-peptide presentation was due mainly to reduced supply of peptides owing to an inhibition of overall protein synthesis. Consequently, generation of H2Kb-SIINFEKL complexes was curtailed during ER stress, illustrating how generation of MHC I peptide ligands is tightly coupled to ongoing protein synthesis. Notably, the UPR-induced decline of MHC I-peptide presentation was more severe when the protein source of peptides was localized in the cytosol than in the ER. This difference was not due to changes in the translation rates of the precursor proteins but to increased stability of the cytosolic protein during ER stress. Conclusion Our results demonstrate that ER stress impairs MHC I-peptide presentation, and that it differentially regulates expression of ER- vs. cytosol-derived peptides. Furthermore, this work illustrates how ER stress, a typical feature of infected and malignant cells, can impinge on cues for adaptive immune recognition.
Collapse
Affiliation(s)
- Diana P Granados
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada.
| | | | | | | | | | | | | |
Collapse
|
33
|
Suzuki T, Sawaki D, Aizawa K, Munemasa Y, Matsumura T, Ishida J, Nagai R. Kruppel-like factor 5 shows proliferation-specific roles in vascular remodeling, direct stimulation of cell growth, and inhibition of apoptosis. J Biol Chem 2009; 284:9549-57. [PMID: 19189969 DOI: 10.1074/jbc.m806230200] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Krüppel-like factor 5 (KLF5), originally isolated as a regulator of phenotypic modulation of vascular smooth muscle cells, induces pathological cell growth and is expressed in the neointima. Although induction of KLF5 up-regulates growth factors like platelet-derived growth factor-A chain, how KLF5 actually contributes to vascular remodeling, notably its direct effects on cell proliferation, had been poorly clarified. To investigate the effects of KLF5 on neointimal formation, we at first performed adenoviral overexpression of KLF5 to rats subjected to carotid balloon injury. Neointimal formation and proliferating cell nuclear antigen-positive rate were significantly increased at 14 days after injury in the KLF5-treated animals. At the cellular level, overexpression of KLF5 also resulted in markedly increased cell proliferation and cell cycle progression. As a molecular mechanism, we showed that KLF5 directly bound to the promoter and up-regulated gene expression of cyclin D1, as well as showing specific transactivation of cyclins and cyclin-dependent kinase inhibitors in cardiovascular cells. Conversely, knockdown of KLF5 by RNA interference specifically down-regulated cyclin D1 and impaired vascular smooth muscle cell proliferation. Furthermore, KLF5 attenuated cleavage of caspase-3 under conditions of apoptotic stimulation. Moreover, KLF5-administered animals exhibited a significant decrease in terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling-positive cells in the medial layer, suggesting inhibition of apoptosis in the early phase after denudation. These findings collectively suggest that KLF5 plays a central role in cardiovascular pathologies through direct and specific stimulation of cell growth as well as inhibition of apoptosis.
Collapse
Affiliation(s)
- Toru Suzuki
- Department of Cardiovascular Medicine and Ubiquitous Preventive Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | | | | | | | | | |
Collapse
|
34
|
Minta JO, Yun JJ, Kabiawu O, Jones J. mRNA differential display identification of vascular smooth muscle early response genes regulated by PDGF. Mol Cell Biochem 2009; 281:63-75. [PMID: 16328958 DOI: 10.1007/s11010-006-0524-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Accepted: 06/29/2005] [Indexed: 11/28/2022]
Abstract
The modulation of vascular smooth muscle cells (VSMCs) from the quiescent phenotype to the proliferative and migratory phenotype is a critical event in the pathogenesis of atherosclerosis. To-date several growth factors, including platelet-derived growth factor, PDGF, have been shown to induce VSMC proliferation and migration. To further understand the mechanism of PDGF-induced VSMC activation, quiescent human coronary artery SMC were treated with PDGF, and the genes that displayed transcriptional changes within 3 and 8 h were identified using differential display RT-PCR, real-time PCR, nucleotide sequencing and bioinformatics. Eleven genes that were highly upregulated or down-regulated at 3 and/or 8 h by PDGF, designated growth-factor regulated VSMC genes (GRSG1-11), were analyzed. GRSG5 and GRSG9-1 were identified as cortactin and cytochrome c oxidase subunit II, respectively. The remaining nine GRSGs were novel. GRSG3, 4, 5 and 9-2 showed wide tissue distribution whereas GRSG10-1, 10-2, and 11 were tissue specific. Cortactin was localized by immunohistochemical staining to the neointima and fibrous cap of human coronary artery atherosclerotic plaques. Domain analysis of open reading frames suggest that the novel GRSGs may participate in signaling, metabolic, translational or migrational processes during PDGF-induced VSMC activation.
Collapse
Affiliation(s)
- Joe O Minta
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada M5S 1A8.
| | | | | | | |
Collapse
|
35
|
Voisin L, Julien C, Duhamel S, Gopalbhai K, Claveau I, Saba-El-Leil MK, Rodrigue-Gervais IG, Gaboury L, Lamarre D, Basik M, Meloche S. Activation of MEK1 or MEK2 isoform is sufficient to fully transform intestinal epithelial cells and induce the formation of metastatic tumors. BMC Cancer 2008; 8:337. [PMID: 19014680 PMCID: PMC2596176 DOI: 10.1186/1471-2407-8-337] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Accepted: 11/17/2008] [Indexed: 12/30/2022] Open
Abstract
Background The Ras-dependent ERK1/2 MAP kinase signaling pathway plays a central role in cell proliferation control and is frequently activated in human colorectal cancer. Small-molecule inhibitors of MEK1/MEK2 are therefore viewed as attractive drug candidates for the targeted therapy of this malignancy. However, the exact contribution of MEK1 and MEK2 to the pathogenesis of colorectal cancer remains to be established. Methods Wild type and constitutively active forms of MEK1 and MEK2 were ectopically expressed by retroviral gene transfer in the normal intestinal epithelial cell line IEC-6. We studied the impact of MEK1 and MEK2 activation on cellular morphology, cell proliferation, survival, migration, invasiveness, and tumorigenesis in mice. RNA interference was used to test the requirement for MEK1 and MEK2 function in maintaining the proliferation of human colorectal cancer cells. Results We found that expression of activated MEK1 or MEK2 is sufficient to morphologically transform intestinal epithelial cells, dysregulate cell proliferation and induce the formation of high-grade adenocarcinomas after orthotopic transplantation in mice. A large proportion of these intestinal tumors metastasize to the liver and lung. Mechanistically, activation of MEK1 or MEK2 up-regulates the expression of matrix metalloproteinases, promotes invasiveness and protects cells from undergoing anoikis. Importantly, we show that silencing of MEK2 expression completely suppresses the proliferation of human colon carcinoma cell lines, whereas inactivation of MEK1 has a much weaker effect. Conclusion MEK1 and MEK2 isoforms have similar transforming properties and are able to induce the formation of metastatic intestinal tumors in mice. Our results suggest that MEK2 plays a more important role than MEK1 in sustaining the proliferation of human colorectal cancer cells.
Collapse
Affiliation(s)
- Laure Voisin
- Institut de Recherche en Immunologie et Cancérologie, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Radu M, Soprano DR, Soprano KJ. S10 phosphorylation of p27 mediates atRA induced growth arrest in ovarian carcinoma cell lines. J Cell Physiol 2008; 217:558-68. [PMID: 18615582 PMCID: PMC2575059 DOI: 10.1002/jcp.21532] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
All trans retinoic acid (atRA) has been shown to inhibit the growth of CAOV3 ovarian carcinoma cells and to elevate the level of p27 cyclin-dependent kinase inhibitor. We report here that phosphorylation at S10 residue is an important event in mediating p27 role in atRA induced growth arrest. atRA treatment of atRA sensitive CAOV3 cells increases the levels of S10 phospho-p27 in both nuclear and cytoplasmic cell compartments. This increase is accompanied by a decrease in the levels of skp2 protein. This effect was not observed in SKOV3 cells which are resistant to atRA growth inhibitory effect. An A10-p27 mutant that cannot be phosphorylated at S10 induces a dominant negative effect on the atRA effect on the levels and activity of endogenous p27. Overexpression of A10-p27 mutant renders CAOV3 cells more resistant to atRA treatment and reverses the effect that atRA has on p27 binding to CDKs, on CDK activity, and on the expression of S phase genes.
Collapse
Affiliation(s)
- Maria Radu
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
37
|
Déléris P, Rousseau J, Coulombe P, Rodier G, Tanguay PL, Meloche S. Activation loop phosphorylation of the atypical MAP kinases ERK3 and ERK4 is required for binding, activation and cytoplasmic relocalization of MK5. J Cell Physiol 2008; 217:778-88. [PMID: 18720373 DOI: 10.1002/jcp.21560] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
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.
Collapse
Affiliation(s)
- Paul Déléris
- Institut de Recherche en Immunologie et Cancérologie, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
38
|
Mielke K. Growth-arrest-dependent expression and phosphorylation of p27kip at serine10 is mediated by the JNK pathway in C6 glioma cells. Mol Cell Neurosci 2008; 38:301-11. [DOI: 10.1016/j.mcn.2007.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 12/03/2007] [Accepted: 12/06/2007] [Indexed: 12/22/2022] Open
|
39
|
Kadowaki M, Mizuno S, Demura Y, Ameshima S, Miyamori I, Ishizaki T. Effect of hypoxia and Beraprost sodium on human pulmonary arterial smooth muscle cell proliferation: the role of p27kip1. Respir Res 2007; 8:77. [PMID: 17974037 PMCID: PMC2164950 DOI: 10.1186/1465-9921-8-77] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2007] [Accepted: 11/01/2007] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Hypoxia induces the proliferation of pulmonary arterial smooth muscle cell (PASMC) in vivo and in vitro, and prostacyclin analogues are thought to inhibit the growth of PASMC. Previous studies suggest that p27kip1, a kind of cyclin-dependent kinase inhibitor, play an important role in the smooth muscle cell proliferation. However, the mechanism of hypoxia and the subcellular interactions between p27kip1 and prostacyclin analogues in human pulmonary arterial smooth muscle cell (HPASMC) are not fully understood. METHODS We investigated the role of p27kip1 in the ability of Beraprost sodium (BPS; a stable prostacyclin analogue) to inhibit the proliferation of HPASMC during hypoxia. To clarify the biological effects of hypoxic air exposure and BPS on HPASMC, the cells were cultured in a hypoxic chamber under various oxygen concentrations (0.1-21%). Thereafter, DNA synthesis was measured as bromodeoxyuridine (BrdU) incorporation, the cell cycle was analyzed by flow cytometry with propidium iodide staining. The p27kip1 mRNA and protein expression and it's stability was measured by real-time RT-PCR and Western blotting. Further, we assessed the role of p27kip1 in HPASMC proliferation using p27kip1 gene knockdown using small interfering RNA (siRNA) transfection. RESULTS Although severe hypoxia (0.1% oxygen) suppressed the proliferation of serum-stimulated HPASMC, moderate hypoxia (2% oxygen) enhanced proliferation in accordance with enhanced p27kip1 protein degradation, whereas BPS suppressed HPASMC proliferation under both hypoxic and normoxic conditions by suppressing p27kip1 degradation with intracellular cAMP-elevation. The 8-bromo-cyclic adenosine monophosphate (8-Br-cAMP), a cAMP analogue, had similar action as BPS in the regulation of p27kip1. Moderate hypoxia did not affect the stability of p27kip1 protein expression, but PDGF, known as major hypoxia-induced growth factors, significantly decreased p27kip1 protein stability. We also demonstrated that BPS and 8-Br-cAMP suppressed HPASMC proliferation under both hypoxic and normoxic conditions by blocking p27kip1 mRNA degradation. Furthermore, p27kip1 gene silencing partially attenuated the effects of BPS and partially restored hypoxia-induced proliferation. CONCLUSION Our study suggests that moderate hypoxia induces HPASMC proliferation, which is partially dependent of p27kip1 down-regulation probably via the induction of growth factors such as PDGF, and BPS inhibits both the cell proliferation and p27kip1 mRNA degradation through cAMP pathway.
Collapse
Affiliation(s)
- Maiko Kadowaki
- Third Department of Internal Medicine, University of Fukui, 23-3 Eiheiji-cho, Matsuoka, Yoshida-gun, Fukui, Japan.
| | | | | | | | | | | |
Collapse
|
40
|
Hirano M, Kanaide H, Hirano K. Rac1-dependent transcriptional up-regulation of p27Kip1 by homophilic cell-cell contact in vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1500-10. [PMID: 17868934 DOI: 10.1016/j.bbamcr.2007.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 07/18/2007] [Accepted: 08/01/2007] [Indexed: 01/24/2023]
Abstract
The mechanism for the transcriptional up-regulation of p27Kip1 due to the formation of the cell-cell contact was investigated in vascular endothelial cells. The induction of the cell-cell contact by adding an extra number of endothelial cells activated Rac1, up-regulated p27Kip1 mRNA and protein, and also facilitated the cell cycle arrest. Transduction of the Rac1 inhibitor protein using the cell-penetrating peptide or treatment with a Rac1 inhibitor NSC23766 inhibited the p27Kip1 up-regulation and delayed the cell cycle arrest. Rac1 was therefore suggested to mediate the contact-induced transcriptional up-regulation of p27Kip1. The role of Rac1 in the regulation of the p27Kip1 promoter activity was next examined with a luciferase reporter assay. The promoter activity was increased by inducing the cell-cell contact, which was significantly inhibited by the Rac1 inhibitory protein and NSC23766. The evaluation of various truncated promoter regions determined region -620 to -573 nucleotides from the initiation codon to be responsible for the contact-induced, Rac1-dependent activation of the p27Kip1 promoter. The present study thus demonstrated for the first time that the activation of Rac1 due to the cell-cell contact plays a critical role in the transcriptional up-regulation of p27Kip1 in vascular endothelial cells.
Collapse
Affiliation(s)
- Mayumi Hirano
- Division of Molecular Cardiology, Research Institute of Angiocardiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | | | |
Collapse
|
41
|
Auld CA, Fernandes KM, Morrison RF. Skp2-mediated p27(Kip1) degradation during S/G2 phase progression of adipocyte hyperplasia. J Cell Physiol 2007; 211:101-11. [PMID: 17096381 DOI: 10.1002/jcp.20915] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
p27(Kip1), an important regulator of Cdk2 activity and G1/S transition, is tightly regulated in a cell-type and condition-specific manner to integrate mitogenic and differentiation signals governing cell cycle progression. We show that p27 protein levels progressively declined from mid-G1 through late-G2 phase as density-arrested 3T3-L1 preadipocytes synchronously reentered the cell cycle during early stages of adipocyte differentiation. This dramatic fall in p27 protein accumulation was due, at least in part, to a decrease in protein stability. Specific inhibitors of the 26S proteasome were shown to completely block the decrease in p27 protein levels throughout G1, increase the abundance of ubiquitylated p27 protein, and inhibit G1/S transition resulting in G1 arrest. It is further demonstrated that p27 was phosphorylated on threonine 187 during S phase progression by Cdk2 and that phosphorylated p27 was polyubiquitylated and degraded. Furthermore, we demonstrate that Skp2 and Cks1 dramatically increased during S/G2 phase progression concomitantly with the maximal fall in p27 protein. Complete knockdown of Skp2 with RNA interference partially prevented p27 degradation equivalent to that observed with Cdk2 blockade suggesting that the SCF(Skp2) E3 ligase and other proteasome-dependent mechanisms contribute to p27 degradation during preadipocyte replication. Interestingly, Skp2-mediated p27 degradation was not essential for G1/S or S/G2 transition as preadipocytes shifted from quiescence to proliferation during adipocyte hyperplasia. Finally, evidence is presented suggesting that elevated p27 protein in the absence of Skp2 was neutralized by sequestration of p27 protein into Cyclin D1/Cdk4 complexes.
Collapse
Affiliation(s)
- Corinth A Auld
- Department of Nutrition, The University of North Carolina at Greensboro, Greensboro, North Carolina 27402, USA
| | | | | |
Collapse
|
42
|
Chassot AA, Turchi L, Virolle T, Fitsialos G, Batoz M, Deckert M, Dulic V, Meneguzzi G, Buscà R, Ponzio G. Id3 is a novel regulator of p27kip1 mRNA in early G1 phase and is required for cell-cycle progression. Oncogene 2007; 26:5772-83. [PMID: 17404577 DOI: 10.1038/sj.onc.1210386] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
P27kip is a key inhibitory protein of the cell-cycle progression, which is rapidly downregulated in early G1 phase by a post-translational mechanism involving the proteosomal degradation. In this study, using a wounding model that induces cell-cycle entry of human dermal fibroblasts, we demonstrate that p27mRNA is downregulated when cells progress into the G1 phase, and then it returns to its basal level when cells approach the S phase. By using a quantitative polymerase chain reaction screening we identified inhibitors of differentiation (Id3), a bHLH transcriptional repressor, as a candidate mediator accounting for p27 mRNA decrease. Id3 silencing, using an small interfering RNA approach, reversed the injury mediated p27 downregulation demonstrating that Id3 is involved in the transcriptional repression of p27. Reporter gene experiments and a chromatin immunoprecipitation assay showed that Id3 likely exerts its repressive action through ELK1 inhibition. By inhibiting early p27 downregulation, Id3 depletion blocked (i) the G1-phase progression as assessed by the inhibition of pRb phosphorylation and p130 degradation and (ii) the G1/S transition as observed by the inhibition of cyclin A induction, demonstrating that p27 mRNA decrease is required for cell proliferation. Apart from its effect on the early p27 diminution, Id3 appears also involved in the control of the steady-state level of p27 at the G1/S boundary. In conclusion, this study identifies a novel mechanism of p27 regulation which besides p27 protein degradation also implicates a transcriptional mechanism mediated by Id3.
Collapse
Affiliation(s)
- A-A Chassot
- INSERM U634; Faculté de Médecine, Université Nice Sophia Antipolis, Nice cedex, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hinrichsen R, Haunsø S, Busk PK. Different regulation of p27 and Akt during cardiomyocyte proliferation and hypertrophy. Growth Factors 2007; 25:132-40. [PMID: 17852410 DOI: 10.1080/08977190701549835] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Postnatal cardiomyocytes normally grow by hypertrophy but show a limited proliferate response to certain stimuli. Although the proliferative capacity declines shortly after birth, neonatal cardiomyocytes can grow both by hypertrophy and by proliferation. Therefore, we have used neonatal cardiomyocytes to investigate the molecular differences between hypertrophic and proliferative growth of cardiomyocytes. Stimulation of neonatal cardiomyocytes with angiotensin II mainly induced hypertrophy, whereas PDGF only had a minor effect on the size of the myocytes. In contrast, PDGF induced significant proliferation in the cardiomyocyte cultures whereas angiotensin II treatment only resulted in a small increase in the number of cells. Measurement of cyclin D-dependent kinase specific phosphorylation of pRb by immunohistochemistry showed that, both stimuli activate the G1 phase of the cell cycle. By western blotting we found that PDGF-induced proliferation correlates with activation of Akt, inactivation of GSK-3beta and downregulation of the cyclin-dependent kinase inhibitor p27, whereas angiotensin II only had a small effect on Akt, GSK-3beta and p27. Our data support the hypothesis that, the hypertrophic and proliferative responses are both activated by G1 cell cycle molecules. The difference between the two responses appears to be that high amounts of p27 are present during hypertrophic growth, whereas proliferation involves downregulation of p27 and GSK-3beta activity and upregulation of Akt.
Collapse
Affiliation(s)
- Rebecca Hinrichsen
- Cell Biology, Biosystems Department, Risø National Laboratory, DK-4000 Roskilde, Denmark.
| | | | | |
Collapse
|
44
|
Rachdi L, Balcazar N, Elghazi L, Barker DJ, Krits I, Kiyokawa H, Bernal-Mizrachi E. Differential effects of p27 in regulation of beta-cell mass during development, neonatal period, and adult life. Diabetes 2006; 55:3520-8. [PMID: 17130500 DOI: 10.2337/db06-0861] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
beta-Cell cycle progression and proliferation are critical to maintain beta-cell mass in adult mice. Of the cell cycle inhibitors, p27Kip1 is thought to be the primary modulator of the proliferative status in most cell types. p27 plays a role in beta-cell adaptation in genetic models of insulin resistance. To study the role of p27 in beta-cells during physiological conditions and at different stages of beta-cell differentiation, we studied mice deficient of or overexpressing p27. Experiments in p27-deficient mice showed improved glucose tolerance and hyperinsulinemia. These changes were associated with increased islet mass and proliferation. The experiments overexpressing p27 in beta-cells were performed using a doxycycline-inducible model. Interestingly, overexpression of p27 for 16 weeks in beta-cells from adult mice had no effect on glucose tolerance, beta-cell mass, or proliferation. In contrast, induction of p27 expression during beta-cell development or early neonatal period resulted in severe glucose intolerance and reduced beta-cell mass by decreased proliferation. These changes were reversible upon discontinuation of doxycycline. These experiments suggest that p27 is a critical molecule for beta-cell proliferation during beta-cell development and early postnatal life but not for maintenance of adult mass.
Collapse
Affiliation(s)
- Latif Rachdi
- Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8127, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Sànchez-Tilló E, Wojciechowska M, Comalada M, Farrera C, Lloberas J, Celada A. Cyclophilin A is required for M-CSF-dependent macrophage proliferation. Eur J Immunol 2006; 36:2515-24. [PMID: 16909430 DOI: 10.1002/eji.200535270] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immunosuppressor sanglifehrin A (SfA) is a member of a family of immunophilin cyclophilin A-binding molecules and does not inhibit calcineurin activity. Sanglifehrin A inhibits M-CSF-dependent macrophage proliferation by arresting the G1 phase of the cell cycle but does not affect cell viability. This immunosuppressor exerts its action on proliferation by inactivating cyclin-dependent kinase 2 (Cdk2) activity. Moreover, c-myc expression is also repressed. In the early steps of M-CSF signaling, SfA inhibits the phosphorylation of Raf-1 and the external regulated kinases (ERK)1/2 and mitogen-activated protein kinase phosphatase-1, which are required for proliferation. The effects of SfA are not related to a block of the proteosome activity. These data show that immunophilin contributes to M-CSF-dependent proliferation through activation of the Raf-1/MEK/ERK pathway and the regulation of Cdk activities, which is required for cell cycle progression.
Collapse
Affiliation(s)
- Ester Sànchez-Tilló
- Macrophage Biology Group, Institute for Research in Biomedicine, Barcelona Science Park, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
46
|
Sanz-González SM, Melero-Fernández de Mera R, Malek NP, Andrés V. Atheroma development in apolipoprotein E-null mice is not regulated by phosphorylation of p27(Kip1) on threonine 187. J Cell Biochem 2006; 97:735-43. [PMID: 16229012 DOI: 10.1002/jcb.20680] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Excessive cellular proliferation is thought to contribute to neointimal lesion development during atherosclerosis and restenosis after angioplasty. Inhibition of cyclin-dependent kinase (CDK) activity by p27 inhibits mammalian cell growth. Mounting evidence indicates that p27 negatively regulates neointimal thickening in animal models of restenosis and atherosclerosis, and its expression in human neointimal lesions is consistent with such a protective role. Cell cycle progression is facilitated by cyclinE/CDK2-dependent phosphorylation of p27 on threonine 187 (T187) during late G1. The purpose of this study was to assess whether this phosphorylation event plays a role during atherosclerosis. To this end, we generated apolipoprotein E-null mice with both p27 alleles replaced by a mutated form non-phosphorylatable at T187 (apoE-/-p27T187A mice) and investigated the kinetics of atheroma development in these animals compared to apoE-/- controls with an intact p27 gene. Fat feeding resulted in comparable level of hypercholesterolemia in both groups of mice. Surprisingly, aortic p27 expression was not increased in fat-fed apoE-/-p27T187A mice compared with apoE-/- controls. Moreover, atheroma size, lesion cellularity, proliferation, and apoptotic rates were undistinguishable in both groups of fat-fed mice. Thus, in contrast to previous studies that highlight the importance of p27 phosphorylation at T187 on the control of p27 expression and function in different tissues and pathophysiological scenarios, our findings demonstrate that this phosphorylation event is not implicated in the control of aortic p27 expression and atheroma progression in hypercholesterolemic mice.
Collapse
Affiliation(s)
- Silvia M Sanz-González
- Laboratory of Vascular Biology, Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | | | | | | |
Collapse
|
47
|
Akashiba H, Matsuki N, Nishiyama N. Calpain activation is required for glutamate-induced p27 down-regulation in cultured cortical neurons. J Neurochem 2006; 99:733-44. [PMID: 16824045 DOI: 10.1111/j.1471-4159.2006.04100.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Recent evidence suggests that cell cycle-related molecules play pivotal roles in multiple forms of cell death in post-mitotic neurons. Nevertheless, it remains unclear what molecular mechanisms are involved in the regulation of expression levels and activities of these molecules. We showed previously that treatment with extracellular glutamate decreases cyclin-dependent kinase inhibitor p27 before neuronal cell death. In this study, we demonstrate that reductions of both p27 and neuronal viability were dependent on activity of calpain, a Ca(2+)-dependent protease, but not on activity of caspase 3. Interestingly, the glutamate-induced reduction of p27 was not dependent on the ubiquitin-proteasome system. In fact, p27 was present only in the neuronal nucleus, whereas calpain 1, a ubiquitous calpain, was observed both in the neuronal nucleus and cytoplasm in control cultures. Glutamate treatment did not change the localization patterns of p27 and calpain 1. It reduced p27 expression level in the nucleus in a calpain-dependent manner. In vitro experiments using neuronal cell lysate and p27 recombinant protein revealed that p27 was degraded as a substrate of activated calpain 1. These results suggest that calpain(s), activated by glutamate treatment, degrade(s) p27 in the nucleus of neurons, which might promote aberrant cell cycle progression.
Collapse
Affiliation(s)
- Hiroki Akashiba
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
48
|
Lee YS, Liu F, Segil N. A morphogenetic wave of p27Kip1 transcription directs cell cycle exit during organ of Corti development. Development 2006; 133:2817-26. [PMID: 16790479 DOI: 10.1242/dev.02453] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms coordinating cell cycle exit with cell differentiation and organogenesis are a crucial, yet poorly understood, aspect of normal development. The mammalian cyclin-dependent kinase inhibitor p27(Kip1) is required for the correct timing of cell cycle exit in developing tissues, and thus plays a crucial role in this process. Although studies of p27(Kip1) regulation have revealed important posttranscriptional mechanisms regulating p27(Kip1) abundance, little is known about how developmental patterns of p27(Kip1) expression, and thus cell cycle exit, are achieved. Here, we show that during inner ear development transcriptional regulation of p27(Kip1) is the primary determinant of a wave of cell cycle exit that dictates the number of postmitotic progenitors destined to give rise to the hair cells and supporting cells of the organ of Corti. Interestingly, transcriptional induction from the p27(Kip1) gene occurs normally in p27(Kip1)-null mice, indicating that developmental regulation of p27(Kip1) transcription is independent of the timing of cell cycle exit. In addition, cell-type-specific patterns of p27(Kip1) transcriptional regulation are observed in the mature organ of Corti and retina, suggesting that this mechanism is important in differential regulation of the postmitotic state. This report establishes a link between the spatial and temporal pattern of p27(Kip1) transcription and the control of cell number during sensory organ morphogenesis.
Collapse
Affiliation(s)
- Yun-Shain Lee
- Gonda Department of Cell and Molecular Biology, House Ear Institute, 2100 West 3rd Street, Los Angeles, CA 90057, USA
| | | | | |
Collapse
|
49
|
Parmentier JH, Zhang C, Estes A, Schaefer S, Malik KU. Essential role of PKC-zeta in normal and angiotensin II-accelerated neointimal growth after vascular injury. Am J Physiol Heart Circ Physiol 2006; 291:H1602-13. [PMID: 16679391 DOI: 10.1152/ajpheart.01363.2005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The contribution of atypical protein kinase C (PKC)-zeta to ANG II-accelerated restenosis after endoluminal vascular injury was investigated by using the rat carotid balloon injury model. Exposure of injured arteries to ANG II resulted in an extensive neointimal thickening (1.9 times) compared with vehicle at day 14. Treatment with PKC-zeta antisense, but not scrambled, oligonucleotides reduced neointimal formation observed in the presence or absence of ANG II. Examination of early events (2 days) after injury showed an increase in cellularity in the perivascular area of the artery wall that was transferred to the adventitia and media after exposure to ANG II, events blocked by PKC-zeta antisense, but not scrambled, oligonucleotides. A positive correlation between medial cellularity at day 2 and extent of neointimal growth at day 14 was established. Immunohistochemical analysis showed that upregulation of inflammatory markers after injury, as well as infiltration of ED1(+) monocytes/macrophages from the perivascular area to the adventitia, was accelerated by ANG II. However, ANG II-stimulated medial increase in cellularity was proliferation independent, and these cells were monocyte chemoattractant protein-1(+)/vimentin(+) but ED1(-)/VCAM(-). PKC-zeta is degraded after injury, and inhibition of its neosynthesis in medial vascular smooth muscle cells or in infiltrating cells with PKC-zeta antisense attenuated medial cellularity and expression of inflammation mediators without reversing smooth muscle cell dedifferentiation. Together, these data indicate that PKC-zeta plays a critical role in normal and ANG II-accelerated neointimal growth through a mechanism involving upregulation of inflammatory mediators, leading to cell infiltration in the media of the vascular wall.
Collapse
Affiliation(s)
- Jean-Hugues Parmentier
- Dept. of Pharmacology, Crowe Bldg., Rm. 211, Univ. of Tennessee, 874 Union Ave., Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
50
|
González JM, Andrés V. Cytostatic gene therapy for occlusive vascular disease. Expert Opin Ther Pat 2006. [DOI: 10.1517/13543776.16.4.507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|