51
|
Salvi R, Abderrahmani A. Decompensation of β-cells in diabetes: when pancreatic β-cells are on ICE(R). J Diabetes Res 2014; 2014:768024. [PMID: 24672804 PMCID: PMC3941242 DOI: 10.1155/2014/768024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/03/2014] [Indexed: 01/05/2023] Open
Abstract
Insulin production and secretion are temporally regulated. Keeping insulin secretion at rest after a rise of glucose prevents exhaustion and ultimately failure of β-cells. Among the mechanisms that reduce β-cell activity is the inducible cAMP early repressor (ICER). ICER is an immediate early gene, which is rapidly induced by the cyclic AMP (cAMP) signaling cascade. The seminal function of ICER is to negatively regulate the production and secretion of insulin by repressing the genes expression. This is part of adaptive response required for proper β-cells function in response to environmental factors. Inappropriate induction of ICER accounts for pancreatic β-cells dysfunction and ultimately death elicited by chronic hyperglycemia, fatty acids, and oxidized LDL. This review underlines the importance of balancing the negative regulation achieved by ICER for preserving β-cell function and survival in diabetes.
Collapse
Affiliation(s)
- Roberto Salvi
- European Genomic Institute for Diabetes (EGID), Lille 2 University, UMR 8199, 3508 Lille, France
| | - Amar Abderrahmani
- European Genomic Institute for Diabetes (EGID), Lille 2 University, UMR 8199, 3508 Lille, France
- Faculty of Medicine West, 1 Place de Verdun, 59045 Lille, France
| |
Collapse
|
52
|
Lorenz K, Stathopoulou K, Schmid E, Eder P, Cuello F. Heart failure-specific changes in protein kinase signalling. Pflugers Arch 2014; 466:1151-62. [DOI: 10.1007/s00424-014-1462-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 01/19/2014] [Accepted: 01/22/2014] [Indexed: 01/14/2023]
|
53
|
Wu Y, Guo Z, Zhang D, Zhang W, Yan Q, Shi X, Zhang M, Zhao Y, Zhang Y, Jiang B, Cheng T, Bai Y, Wang J. A novel colon cancer gene therapy using rAAV‑mediated expression of human shRNA-FHL2. Int J Oncol 2013; 43:1618-26. [PMID: 24008552 DOI: 10.3892/ijo.2013.2090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/05/2013] [Indexed: 11/06/2022] Open
Abstract
FHL2 (Four and a half LIM-only protein 2) has been identified as an oncogene in colon cancer and suppression of FHL2 induces cell differentiation and tumorigenesis in colon cancer cell lines. The aim of this study was to develop a novel and effective approach to knockdown FHL2, which can serve as a promising target of colon cancer therapy. Recombinant adeno-associated virus (rAAV) was generated bearing with FHL2-shRNA and transfected into LoVo cells. Cell cycle and growth were assessed. The interaction between FHL2 and G0/G1 cell cycle and growth was evaluated by flow cytometry, western blot analysis and WST-1 assay. We showed that suppression of FHL2 by rAAV-shRNA induced G0/G1 cell cycle arrest and inhibited cell growth. Apoptosis-related proteins and their activity was investigated at the same time. rAAV-FHL2‑shRNA activated intrinsic and extrinsic apoptotic pathways and increased cell susceptibility to apoptotic stimuli by 5-FU. Moreover, a xenograft model was established to explore rAAV-FHL2-shRNA with 5-FU mediated tumorigenesis in vivo. A strong anti-tumorigenic effect of rAAV-FHL2-shRNA was shown in nude mice and this antitumor effect was enhanced when combined with 5-FU treatment. These findings implicate FHL2 as a cell cycle and growth modulator and thus inhibit apoptosis in colon cancer cells. rAAV-shRNA-FHL2 may serve as a novel and potent therapeutic or 5-FU co-therapeutic agent for colon cancer.
Collapse
Affiliation(s)
- Yao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Digestive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Anttila V, Winsvold BS, Gormley P, Kurth T, Bettella F, McMahon G, Kallela M, Malik R, de Vries B, Terwindt G, Medland SE, Todt U, McArdle WL, Quaye L, Koiranen M, Ikram MA, Lehtimäki T, Stam AH, Ligthart L, Wedenoja J, Dunham I, Neale BM, Palta P, Hamalainen E, Schürks M, Rose LM, Buring JE, Ridker PM, Steinberg S, Stefansson H, Jakobsson F, Lawlor DA, Evans DM, Ring SM, Färkkilä M, Artto V, Kaunisto MA, Freilinger T, Schoenen J, Frants RR, Pelzer N, Weller CM, Zielman R, Heath AC, Madden PA, Montgomery GW, Martin NG, Borck G, Göbel H, Heinze A, Heinze-Kuhn K, Williams FM, Hartikainen AL, Pouta A, van den Ende J, Uitterlinden AG, Hofman A, Amin N, Hottenga JJ, Vink JM, Heikkilä K, Alexander M, Muller-Myhsok B, Schreiber S, Meitinger T, Wichmann HE, Aromaa A, Eriksson JG, Traynor B, Trabzuni D, North American Brain Expression Consortium, UK Brain Expression Consortium, Rossin E, Lage K, Jacobs SB, Gibbs JR, Birney E, Kaprio J, Penninx BW, Boomsma DI, van Duijn C, Raitakari O, Jarvelin MR, Zwart JA, Cherkas L, Strachan DP, Kubisch C, Ferrari MD, van den Maagdenberg AM, Dichgans M, Wessman M, Smith GD, Stefansson K, Daly MJ, Nyholt DR, Chasman D, Palotie A. Genome-wide meta-analysis identifies new susceptibility loci for migraine. Nat Genet 2013; 45:912-917. [PMID: 23793025 PMCID: PMC4041123 DOI: 10.1038/ng.2676] [Citation(s) in RCA: 298] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 05/30/2013] [Indexed: 12/15/2022]
Abstract
Migraine is the most common brain disorder, affecting approximately 14% of the adult population, but its molecular mechanisms are poorly understood. We report the results of a meta-analysis across 29 genome-wide association studies, including a total of 23,285 individuals with migraine (cases) and 95,425 population-matched controls. We identified 12 loci associated with migraine susceptibility (P<5×10(-8)). Five loci are new: near AJAP1 at 1p36, near TSPAN2 at 1p13, within FHL5 at 6q16, within C7orf10 at 7p14 and near MMP16 at 8q21. Three of these loci were identified in disease subgroup analyses. Brain tissue expression quantitative trait locus analysis suggests potential functional candidate genes at four loci: APOA1BP, TBC1D7, FUT9, STAT6 and ATP5B.
Collapse
Affiliation(s)
- Verneri Anttila
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bendik S. Winsvold
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Department of Neurology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Padhraig Gormley
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | - Tobias Kurth
- INSERM Unit 708 – Neuroepidemiology, F-33000 Bordeaux, France
- University of Bordeaux, F-33000 Bordeaux, France
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | | | - George McMahon
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Mikko Kallela
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Rainer Malik
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
| | - Boukje de Vries
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gisela Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sarah E. Medland
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Unda Todt
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Wendy L. McArdle
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Lydia Quaye
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Markku Koiranen
- Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - M. Arfan Ikram
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Radiology Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Neurology Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Anine H. Stam
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lannie Ligthart
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
- EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Juho Wedenoja
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
| | - Ian Dunham
- European Bioinformatics Insitute, Wellcome Trust Genome Campus, Cambridge, UK
| | - Benjamin M. Neale
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Priit Palta
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Eija Hamalainen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Markus Schürks
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Lynda M Rose
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Julie E. Buring
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Paul M. Ridker
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
- Harvard Medical School, Boston, MA 02215, USA
| | | | | | - Finnbogi Jakobsson
- Department of Neurology, Landspitali University Hospital, Reykjavik, Iceland
| | - Debbie A. Lawlor
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - David M. Evans
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Susan M. Ring
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Markus Färkkilä
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Ville Artto
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Mari A Kaunisto
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Tobias Freilinger
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Neurology, Klinikum der Universität München, Munich, Germany
| | - Jean Schoenen
- Headache Research Unit, Department of Neurology and Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-Neurosciences, Liège University, Liège, Belgium
| | - Rune R. Frants
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Nadine Pelzer
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Claudia M. Weller
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ronald Zielman
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pamela A.F. Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Nicholas G. Martin
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Guntram Borck
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | | | - Axel Heinze
- Kiel Pain and Headache Center, Kiel, Germany
| | | | - Frances M.K. Williams
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Anna-Liisa Hartikainen
- Department of Clinical Sciences/Obstetrics and Gynecology, University Hospital of Oulu, Oulu, Finland
| | - Anneli Pouta
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Department of Clinical Sciences/Obstetrics and Gynecology, University Hospital of Oulu, Oulu, Finland
- Department of Children, Young People and Families, National Institute for Health and Welfare, Helsinki, Finland
| | - Joyce van den Ende
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Albert Hofman
- Genetic Epidemiology Unit, Department of Clinical Genetics, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Jacqueline M. Vink
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Kauko Heikkilä
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
| | - Michael Alexander
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Bertram Muller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan Schreiber
- Department of Clinical Molecular Biology, Christian Albrechts University, Kiel, Germany
- Department of Internal Medicine I, Christian Albrechts University, Kiel, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Human Genetics, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Heinz Erich Wichmann
- Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Ludwig-Maximilians-Universität München, Munich, Germany
- Institute of Epidemiology I, HelmholtzCenter Munich, Neuherberg, Germany
- Klinikum Großhadern, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Arpo Aromaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Johan G. Eriksson
- Folkhälsan Research Center, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice, Helsinki University Central Hospital, Helsinki, Finland
- Vaasa Central Hospital, Vaasa, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Bryan Traynor
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Daniah Trabzuni
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | | | - Elizabeth Rossin
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Kasper Lage
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital for Children, Massachusetts General Hospital, Boston, MA, USA
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
- Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Suzanne B.R. Jacobs
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - J. Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Ewan Birney
- European Bioinformatics Insitute, Wellcome Trust Genome Campus, Cambridge, UK
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, Hjelt Institute, University of Helsinki, Helsinki, Finland
- Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland
| | - Brenda W. Penninx
- EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Department of Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Cornelia van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, University of Oulu, Oulu, Finland
- Department of Children, Young People and Families, National Institute for Health and Welfare, Helsinki, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, MRC-HPA Centre for Environment and Health, Faculty of Medicine, Imperial College, London, UK
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - John-Anker Zwart
- Department of Neurology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Lynn Cherkas
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - David P. Strachan
- Division of Population Health Sciences and Education, St George’s, University of London, London, UK
| | | | - Michel D. Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Arn M.J.M. van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Maija Wessman
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - George Davey Smith
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Kari Stefansson
- deCODE genetics, Reykjavik, Iceland
- School of Medicine, University of Iceland, Reykjavik, Iceland
| | - Mark J. Daly
- Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dale R. Nyholt
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Daniel Chasman
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Aarno Palotie
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
55
|
Soreq L, Salomonis N, Bronstein M, Greenberg DS, Israel Z, Bergman H, Soreq H. Small RNA sequencing-microarray analyses in Parkinson leukocytes reveal deep brain stimulation-induced splicing changes that classify brain region transcriptomes. Front Mol Neurosci 2013; 6:10. [PMID: 23717260 PMCID: PMC3652308 DOI: 10.3389/fnmol.2013.00010] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 04/16/2013] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are key post transcriptional regulators of their multiple target genes. However, the detailed profile of miRNA expression in Parkinson's disease, the second most common neurodegenerative disease worldwide and the first motor disorder has not been charted yet. Here, we report comprehensive miRNA profiling by next-generation small-RNA sequencing, combined with targets inspection by splice-junction and exon arrays interrogating leukocyte RNA in Parkinson's disease patients before and after deep brain stimulation (DBS) treatment and of matched healthy control volunteers (HC). RNA-Seq analysis identified 254 miRNAs and 79 passenger strand forms as expressed in blood leukocytes, 16 of which were modified in patients pre-treatment as compared to HC. 11 miRNAs were modified following brain stimulation 5 of which were changed inversely to the disease induced changes. Stimulation cessation further induced changes in 11 miRNAs. Transcript isoform abundance analysis yielded 332 changed isoforms in patients compared to HC, which classified brain transcriptomes of 47 PD and control independent microarrays. Functional enrichment analysis highlighted mitochondrion organization. DBS induced 155 splice changes, enriched in ubiquitin homeostasis. Cellular composition analysis revealed immune cell activity pre and post treatment. Overall, 217 disease and 74 treatment alternative isoforms were predictably targeted by modified miRNAs within both 3' and 5' untranslated ends and coding sequence sites. The stimulation-induced network sustained 4 miRNAs and 7 transcripts of the disease network. We believe that the presented dynamic networks provide a novel avenue for identifying disease and treatment-related therapeutic targets. Furthermore, the identification of these networks is a major step forward in the road for understanding the molecular basis for neurological and neurodegenerative diseases and assessment of the impact of brain stimulation on human diseases.
Collapse
Affiliation(s)
- Lilach Soreq
- Department of Medical Neurobiology, Hadassah Faculty of Medicine, The Hebrew University of JerusalemJerusalem, Israel
| | - Nathan Salomonis
- Division of Genomic Medicine, The Gladstone Institute of Cardiovascular DiseaseSan Francisco, CA, USA
| | - Michal Bronstein
- The Center for Genomic Technologies, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - David S. Greenberg
- Department of Biological Chemistry, The Life Sciences Institute, The Hebrew University of JerusalemJerusalem, Israel
| | - Zvi Israel
- Department of Neurosurgery, The Centre for Functional and Restorative Neurosurgery, Hadassah University HospitalJerusalem, Israel
| | - Hagai Bergman
- Department of Medical Neurobiology, Hadassah Faculty of Medicine, The Hebrew University of JerusalemJerusalem, Israel
- The Edmond and Lili Safra Center for Brain Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Hermona Soreq
- Department of Biological Chemistry, The Life Sciences Institute, The Hebrew University of JerusalemJerusalem, Israel
- The Edmond and Lili Safra Center for Brain Sciences, The Hebrew University of JerusalemJerusalem, Israel
| |
Collapse
|
56
|
The four and a half LIM family members are novel interactants of the human T-cell leukemia virus type 1 Tax oncoprotein. J Virol 2013; 87:7435-44. [PMID: 23616667 DOI: 10.1128/jvi.00070-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the etiologic agent of adult T-cell leukemia (ATL). The viral regulatory protein Tax1 plays a pivotal role in T-cell transformation and ATL development. Previous studies in our laboratory, using the yeast 2-hybrid approach to screen a T-cell library for Tax1-interacting partners, identified the cellular Four and a Half Lim domain protein 3 (FHL3) as a possible Tax1-interacting candidate. FHL3 is a member of the FHL family of proteins, which function as transcriptional coactivators and cytoskeleton regulators and have a role in cancer progression and development. The aim of this study was to investigate the physical and functional interaction between Tax1 and members of the FHL family of proteins. We show that Tax1 and FHL3 interact both in vitro and in vivo. Furthermore, both FHL1 and -2 also interact with Tax1. We have demonstrated that FHL3 enhances Tax1-mediated activation of the viral long terminal repeat (LTR) without affecting basal activity and that FHL1 to -3 regulate NF-κB activation by Tax1 in a cell-specific manner. In addition, we have found that the interaction between Tax1 and FHL1 to -3 affects the localization of these proteins, leading to their redistribution in cells. Tax1 also affected FHL3 cytoskeleton function by increasing FHL3-mediated cell spreading. Overall, our results suggest that the interaction between Tax1 and the FHL family alters both the transactivating activity and the subcellular localization of Tax1 and provide new insights into molecular mechanisms that underlie the oncogenic nature of this HTLV-1 protein.
Collapse
|
57
|
Xia T, Lévy L, Levillayer F, Jia B, Li G, Neuveut C, Buendia MA, Lan K, Wei Y. The four and a half LIM-only protein 2 (FHL2) activates transforming growth factor β (TGF-β) signaling by regulating ubiquitination of the E3 ligase Arkadia. J Biol Chem 2012; 288:1785-94. [PMID: 23212909 DOI: 10.1074/jbc.m112.439760] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Arkadia is a RING-based ubiquitin ligase that positively regulates TGF-β signaling by targeting several pathway components for ubiquitination and degradation. However, little is known about the mechanisms controlling Arkadia activity. Here we show that the LIM-only protein FHL2 binds and synergistically cooperates with Arkadia to activate Smad3/Smad4-dependent transcription. Knockdown of FHL2 by RNA interference decreases Arkadia level and restricts the amplitude of Arkadia-induced TGF-β target gene responses. We found that Arkadia is ubiquitinated via K63- and K27-linked polyubiquitination. A single mutation at the RING domain that abolishes the E3 activity diminishes Arkadia ubiquitination, indicating that this modification partly involves autocatalytic process. Mutation of seven lysines at the C-terminal region of Arkadia severely impairs ubiquitination through the K27 but not the K63 linkage and slows down the turnover of Arkadia, suggesting that K27-linked polyubiquitination might promote proteolysis-dependent regulation of Arkadia. We show that FHL2 increases the half-life of Arkadia through inhibition of ubiquitin chain assembly on the protein, which provides a molecular basis for functional cooperation between Arkadia and FHL2 in enhancing TGF-β signaling. Our study uncovers a novel regulatory mechanism of Arkadia by ubiquitination and identifies FHL2 as important regulator of Arkadia ubiquitination and TGF-β signal transduction.
Collapse
Affiliation(s)
- Tian Xia
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai Institute for Biological Sciences, 225 South Chongqing Road, 200025, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Nakanishi K, Saito Y, Azuma N, Sasajima T. Cyclic adenosine monophosphate response-element binding protein activation by mitogen-activated protein kinase-activated protein kinase 3 and four-and-a-half LIM domains 5 plays a key role for vein graft intimal hyperplasia. J Vasc Surg 2012; 57:182-93, 193.e1-10. [PMID: 23127979 DOI: 10.1016/j.jvs.2012.06.082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 06/07/2012] [Accepted: 06/09/2012] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Intimal hyperplasia (IH) is the main cause of vein graft stenosis or failure after bypass surgery. Basic investigations are proceeding in an animal model of mechanically desquamated arteries, and numerous molecules for potential IH treatments have been identified; however, neither insights into the mechanism of IH nor substantially effective treatments for its suppression have been developed. The goals of the present study are to use human vein graft samples to identify therapeutic target genes that control IH and to investigate the therapeutic efficacy of these candidate molecules in animal models. METHODS Using microarray analysis of human vein graft samples, we identified two previously unrecognized IH-related genes, mitogen-activated protein kinase-activated protein kinase 3 (MAPKAPK3) and four-and-a-half LIM domains 5 (FHL5). RESULTS Transfer of either candidate gene resulted in significantly elevated vascular smooth muscle cell (VSMC) proliferation and migration. Interestingly, cotransfection of both genes increased VSMC proliferation in an additive manner. These genes activated cyclic adenosine monophosphate response-element (CRE) binding protein (CREB), but their mechanisms of activation were different. MAPKAPK3 phosphorylated CREB, but FHL5 bound directly to CREB. A CREB dominant-negative protein, KCREB, which blocks its ability to bind CRE, repressed VSMC proliferation and migration. In a wire-injury mouse model, gene transfer of KCREB plasmid significantly repressed IH. In this vessel tissue, CRE-activated gene expression was repressed. Furthermore, we confirmed the changes in MAPKAPK3 and FHL5 expression using vein graft samples from eight patients. CONCLUSIONS We successively identified two previously unrecognized IH activators, MAPKAPK3 and FHL5, using human vein graft samples. Gene transfer of KCREB repressed IH in an animal model. Inhibition of CREB function is a promising gene therapy strategy for IH.
Collapse
Affiliation(s)
- Keisuke Nakanishi
- Department of Surgery, Asahikawa Medical University, Hokkaido, Japan
| | | | | | | |
Collapse
|
59
|
Nouët Y, Dahan J, Labalette C, Levillayer F, Julien B, Jouvion G, Cairo S, Vives FL, Ribeiro A, Huerre M, Colnot S, Perret C, Nhieu JTV, Tordjmann T, Buendia MA, Wei Y. The four and a half LIM-only protein 2 regulates liver homeostasis and contributes to carcinogenesis. J Hepatol 2012; 57:1029-36. [PMID: 22796152 DOI: 10.1016/j.jhep.2012.06.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS The four and a half LIM-only protein 2 (FHL2) is upregulated in diverse pathological conditions. Here, we analyzed the effects of FHL2 overexpression in the liver of FHL2 transgenic mice (Apo-FHL2). METHODS We first examined cell proliferation and apoptosis in Apo-FHL2 livers and performed partial hepatectomy to investigate high FHL2 expression in liver regeneration. Expression of FHL2 was then analyzed by real time PCR in human hepatocellular carcinoma and adjacent non-tumorous livers. Finally, the role of FHL2 in hepatocarcinogenesis was assessed using Apo-FHL2;Apc(lox/lox) mice. RESULTS Six-fold increase in cell proliferation in transgenic livers was associated with concomitant apoptosis, resulting in normal liver mass. In Apo-FHL2 livers, both cyclin D1 and p53 were markedly increased. Evidence supporting a p53-dependent cell death mechanism was provided by the findings that FHL2 bound to and activated the p53 promoter, and that a dominant negative p53 mutant compromised FHL2-induced apoptosis in hepatic cells. Following partial hepatectomy in Apo-FHL2 mice, hepatocytes displayed advanced G1 phase entry and DNA synthesis leading to accelerated liver weight restoration. Interestingly, FHL2 upregulation in human liver specimens showed significant association with increasing inflammation score and cirrhosis. Finally, while Apo-FHL2 mice developed no tumors, the FHL2 transgene enhanced hepatocarcinogenesis induced by liver-specific deletion of the adenomatous polyposis coli gene and aberrant Wnt/β-catenin signaling in Apc(lox/lox) animals. CONCLUSIONS Our results implicate FHL2 in the regulation of signaling pathways that couple proliferation and cell death machineries, and underscore the important role of FHL2 in liver homeostasis and carcinogenesis.
Collapse
Affiliation(s)
- Yann Nouët
- Institut Pasteur, Unité d'Oncogenèse et Virologie Moléculaire, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Matulis CK, Mayo KE. The LIM domain protein FHL2 interacts with the NR5A family of nuclear receptors and CREB to activate the inhibin-α subunit gene in ovarian granulosa cells. Mol Endocrinol 2012; 26:1278-90. [PMID: 22734036 DOI: 10.1210/me.2011-1347] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nuclear receptor transcriptional activity is enhanced by interaction with coactivators. The highly related nuclear receptor 5A (NR5A) subfamily members liver receptor homolog 1 and steroidogenic factor 1 bind to and activate several of the same genes, many of which are important for reproductive function. To better understand transcriptional activation by these nuclear receptors, we sought to identify interacting proteins that might function as coactivators. The LIM domain protein four and a half LIM domain 2 (FHL2) was identified as interacting with the NR5A receptors in a yeast two-hybrid screen of a human ovary cDNA library. FHL2, and the closely related FHL1, are both expressed in the rodent ovary and in granulosa cells. Small interfering RNA-mediated knockdown of FHL1 and FHL2 in primary mouse granulosa cells reduced expression of the NR5A target genes encoding inhibin-α and P450scc. In vitro assays confirmed the interaction between the FHL and NR5A proteins and revealed that a single LIM domain of FHL2 is sufficient for this interaction, whereas determinants in both the ligand binding domain and DNA binding domain of NR5A proteins are important. FHL2 enhances the ability of both liver receptor homolog 1 and steroidogenic factor 1 to activate the inhibin-α subunit gene promoter in granulosa cells and thus functions as a transcriptional coactivator. FHL2 also interacts with cAMP response element-binding protein and substantially augments activation of inhibin gene expression by the combination of NR5A receptors and forskolin, suggesting that FHL2 may facilitate integration of these two signals. Collectively these results identify FHL2 as a novel coactivator of NR5A nuclear receptors in ovarian granulosa cells and suggest its involvement in regulating target genes important for mammalian reproduction.
Collapse
Affiliation(s)
- Christina K Matulis
- Department of Molecular Biosciences and Center of Reproductive Science, Northwestern University, Evanston, Illinois 60208, USA
| | | |
Collapse
|
61
|
Wong CH, Mak GWY, Li MS, Tsui SKW. The LIM-only protein FHL2 regulates interleukin-6 expression through p38 MAPK mediated NF-κB pathway in muscle cells. Cytokine 2012; 59:286-93. [PMID: 22633286 DOI: 10.1016/j.cyto.2012.04.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 03/23/2012] [Accepted: 04/22/2012] [Indexed: 01/09/2023]
Abstract
Interleukin 6 (IL-6) is pleiotropic cytokine playing an important role in inflammatory response. Other than classical immune tissues, IL-6 is also produced in muscle cells under specific conditions. Four-and-a-half LIM-only protein 2 (FHL2) is preferentially expressed in skeletal and cardiac muscle cells compared to other tissues indicating it has an important role in skeletal muscle and cardiovascular system. In this report, the regulation of IL-6 by FHL2 in muscle cells was investigated. We demonstrated that FHL2 overexpression increased IL-6 mRNA level and its protein secretion in skeletal myoblasts. In contrast, the IL-6 secretion was significantly decreased after FHL2-knockdown by siRNA in response to TNFα stimulation. We further showed that FHL2-mediated induction of IL-6 was regulated by the activation of IL-6 promoter through stimulating NF-κB and p38 MAPK signaling pathway. Our results further illustrated the molecular mechanisms of IL-6 production, which provides new insights in the roles of FHL2 in post-injury inflammation or cytoprotection of muscle cells.
Collapse
Affiliation(s)
- Chi-Hang Wong
- Department of Clinical Oncology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong Special Administrative Region, China
| | | | | | | |
Collapse
|
62
|
Rafael MS, Laizé V, Bensimon-Brito A, Leite RB, Schüle R, Cancela ML. Four-and-a-half LIM domains protein 2 (FHL2) is associated with the development of craniofacial musculature in the teleost fish Sparus aurata. Cell Mol Life Sci 2012; 69:423-34. [PMID: 21739231 PMCID: PMC11115147 DOI: 10.1007/s00018-011-0754-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 06/08/2011] [Accepted: 06/14/2011] [Indexed: 01/08/2023]
Abstract
Four-and-a-half LIM domains protein 2 (FHL2) is involved in major cellular mechanisms such as regulation of gene transcription and cytoskeleton modulation, participating in physiological control of cardiogenesis and osteogenesis. Knowledge on underlying mechanisms is, however, limited. We present here new data on FHL2 protein and its role during vertebrate development using a marine teleost fish, the gilthead seabream (Sparus aurata L.). In silico comparison of vertebrate protein sequences and prediction of LIM domain three-dimensional structure revealed a high degree of conservation, suggesting a conserved function throughout evolution. Determination of sites and levels of FHL2 gene expression in seabream indicated a central role for FHL2 in the development of heart and craniofacial musculature, and a potential role in tissue calcification. Our data confirmed the key role of FHL2 protein during vertebrate development and gave new insights into its particular involvement in craniofacial muscle development and specificity for slow fibers.
Collapse
Affiliation(s)
- Marta S. Rafael
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Vincent Laizé
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anabela Bensimon-Brito
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Ricardo B. Leite
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Roland Schüle
- Department of Urology/Women’s Hospital and Center for Clinical Research, University of Freiburg Medical Center, Breisacherstrasse 66, 79106 Freiburg, Germany
| | - M. Leonor Cancela
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Department of Biomedical Sciences and Medicine (DCBM), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
63
|
Zheng Q, Zhao Y. The diverse biofunctions of LIM domain proteins: determined by subcellular localization and protein-protein interaction. Biol Cell 2012; 99:489-502. [PMID: 17696879 DOI: 10.1042/bc20060126] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The LIM domain is a cysteine- and histidine-rich motif that has been proposed to direct protein-protein interactions. A diverse group of proteins containing LIM domains have been identified, which display various functions including gene regulation and cell fate determination, tumour formation and cytoskeleton organization. LIM domain proteins are distributed in both the nucleus and the cytoplasm, and they exert their functions through interactions with various protein partners.
Collapse
Affiliation(s)
- Quanhui Zheng
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
64
|
Abstract
During the past 2 years, considerable progress in the field of four and a half LIM domain protein 1 (FHL1)-related myopathies has led to the identification of a growing number of FHL1 mutations. This genetic progress has uncovered crucial pathophysiological concepts, thus redefining clinical phenotypes. Important new characterizations include 4 distinct human myopathies: reducing body myopathy, X-linked myopathy with postural muscle atrophy, Emery-Dreifuss muscular dystrophy, and scapuloperoneal myopathy. Additionally, FHL1 mutations have been discovered in rigid spine syndrome and in a single family with contractures, rigid spine, and cardiomyopathy. In this review, we focus on the clinical phenotypes, which we correlate with the novel genetic and histological findings encountered within FHL1-related myopathies. This correlation will frequently lead to a considerably expanded clinical spectrum associated with a given FHL1 mutation.
Collapse
|
65
|
Mitton B, Cho EC, Aldana-Masangkay GI, Sakamoto KM. The function of cyclic-adenosine monophosphate responsive element-binding protein in hematologic malignancies. Leuk Lymphoma 2011; 52:2057-63. [DOI: 10.3109/10428194.2011.584994] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
66
|
Expression of Crip2, a LIM-domain-only protein, in the mouse cardiovascular system under physiological and pathological conditions. Gene Expr Patterns 2011; 11:384-94. [DOI: 10.1016/j.gep.2011.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 05/02/2011] [Accepted: 05/05/2011] [Indexed: 01/08/2023]
|
67
|
Cowling BS, Cottle DL, Wilding BR, D'Arcy CE, Mitchell CA, McGrath MJ. Four and a half LIM protein 1 gene mutations cause four distinct human myopathies: a comprehensive review of the clinical, histological and pathological features. Neuromuscul Disord 2011; 21:237-51. [PMID: 21310615 DOI: 10.1016/j.nmd.2011.01.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 12/20/2010] [Accepted: 01/01/2011] [Indexed: 10/18/2022]
Abstract
Mutations in the four and a half LIM protein 1 (FHL1) gene were recently identified as the cause of four distinct skeletal muscle diseases. Since the initial report outlining the first fhl1 mutation in 2008, over 25 different mutations have been identified in patients with reducing body myopathy, X-linked myopathy characterized by postural muscle atrophy, scapuloperoneal myopathy and Emery-Dreifuss muscular dystrophy. Reducing body myopathy was first described four decades ago, its underlying genetic cause was unknown until the discovery of fhl1 mutations. X-linked myopathy characterized by postural muscle atrophy is a novel disease where fhl1 mutations are the only cause. This review will profile each of the FHL1, with a comprehensive analysis of mutations, a comparison of the clinical and histopathological features and will present several hypotheses for the possible disease mechanism(s).
Collapse
Affiliation(s)
- Belinda S Cowling
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | | | | | | | | | | |
Collapse
|
68
|
Ng CF, Zhou WJW, Ng PKS, Li MS, Ng YK, Lai PBS, Tsui SKW. Characterization of human FHL2 transcript variants and gene expression regulation in hepatocellular carcinoma. Gene 2011; 481:41-7. [DOI: 10.1016/j.gene.2011.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/15/2011] [Indexed: 11/25/2022]
|
69
|
Ding L, Niu C, Zheng Y, Xiong Z, Liu Y, Lin J, Sun H, Huang K, Yang W, Li X, Ye Q. FHL1 interacts with oestrogen receptors and regulates breast cancer cell growth. J Cell Mol Med 2011; 15:72-85. [PMID: 19840196 PMCID: PMC3822495 DOI: 10.1111/j.1582-4934.2009.00938.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Four and a half LIM protein 1 (FHL1) belongs to the Lin-1, Isl-1 and Mec-3 (LIM)-only protein family and plays important roles in muscle growth and carcinogenesis. However, the biological function of FHL1 remains largely unknown. Here, we show that FHL1 physically and functionally interacted with oestrogen receptors (ERs), which are involved in breast cancer development and progression. FHL1 bound specifically to the activation function-1 domain of ER. Physical interaction of FHL1 and ER is required for FHL1 repression of oestrogen-responsive gene transcription. FHL1 affected recruitment of ER to an oestrogen-responsive promoter and ER binding to an oestrogen-responsive element. Overexpression of FHL1 in breast cancer cells decreased expression of oestrogen-responsive proteins, whereas knockdown of endogenous FHL1 with FHL1 small interfering RNA increased the expression of these proteins. Further analysis of 46 breast cancer samples showed that FHL1 expression negatively associated with oestrogen-responsive gene expression in breast cancer cells. FHL1 inhibited anchorage-dependent and -independent breast cancer cell growth. These results suggest that FHL1 may play an important role in ER signalling as well as breast cancer cell growth regulation.
Collapse
Affiliation(s)
- Lihua Ding
- Department of Molecular Oncology, Beijing Institute of Biotechnology, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Sharma P, Shathasivam T, Ignatchenko V, Kislinger T, Gramolini AO. Identification of an FHL1 protein complex containing ACTN1, ACTN4, and PDLIM1 using affinity purifications and MS-based protein-protein interaction analysis. MOLECULAR BIOSYSTEMS 2011; 7:1185-96. [PMID: 21246116 PMCID: PMC3711787 DOI: 10.1039/c0mb00235f] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Four and a half LIM domains protein 1 (FHL1) is the most widely expressed member of the FHL family of proteins, consisting of four and a half highly conserved LIM domains. A multifunctional and integral role for FHL1 has been implicated in muscle development, structural maintenance, and signaling. To date, 27 FHL1 mutations have been identified that result in at least six different X-linked myopathies, with patients often presenting with cardiovascular complications. Since proteins assemble into dynamic complexes within the cell, FHL1 likely mediates its biological functions in conjunction with other proteins. Delineation of FHL1 interactions could provide insight into its regulatory functions. METHODS We performed tandem affinity purification from human embryonic kidney 293 (HEK-293) cells to purify FHL1 and interacting proteins. To identify the potential interactors of FHL1 we performed a total of 9 different purifications from HEK-293 cells which included 3 experimental replicates for each biological condition: FHL1, tag control (DPYSL3), and negative control (empty vector). Purified samples were analyzed by liquid chromatography mass spectrometry (LC-MS). Potential interactors were then verified by immunoprecipitation from mouse heart ventricles and interactions visualized in adult cardiomyocytes using 3D fluorescence microscopy. RESULTS We identified a total of 310 different proteins from all 9 purifications and by applying stringent filtering criteria we eliminated all proteins found in any of the controls and only allowed those that were detected in two or more bait purification. We identified 34 high confidence potential binding partners of FHL1. We then showed that FHL1 exists as part of a complex that binds with PDLIM1, GSN and ACTN1.
Collapse
Affiliation(s)
- Parveen Sharma
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
71
|
Shathasivam T, Kislinger T, Gramolini AO. Genes, proteins and complexes: the multifaceted nature of FHL family proteins in diverse tissues. J Cell Mol Med 2011; 14:2702-20. [PMID: 20874719 PMCID: PMC3822721 DOI: 10.1111/j.1582-4934.2010.01176.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Four and a half LIM domain protein 1 (FHL1) is the founding member of the FHL family of proteins characterized by the presence of four and a half highly conserved LIM domains. The LIM domain is a protein-interaction motif and is involved in linking proteins with both the actin cytoskeleton and transcriptional machinery. To date, more than 25 different protein interactions have been identified for full length FHL1 and its spliced variants, and these interactions can be mapped to a variety of functional classes. Because FHL1 is expressed predominantly in skeletal muscle, all of these proteins interactions translate into a multifunctional and integral role for FHL1 in muscle development, structural maintenance, and signalling. Importantly, 27 FHL1 genetic mutations have been identified that result in at least six different X-linked myopathies, with patients often presenting with cardiovascular disease. FHL1 expression is also significantly up-regulated in a variety of cardiac disorders, even at the earliest stages of disease onset. Alternatively, FHL1 expression is suppressed in a variety of cancers, and ectopic FHL1 expression offers potential for some phenotype rescue. This review focuses on recent studies of FHL1 in muscular dystrophies and cardiovascular disease, and provides a comprehensive review of FHL1s multifunctional roles in skeletal muscle.
Collapse
|
72
|
FHL2 exhibits anti-proliferative and anti-apoptotic activities in liver cancer cells. Cancer Lett 2011; 304:97-106. [PMID: 21377781 DOI: 10.1016/j.canlet.2011.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 01/20/2011] [Accepted: 02/02/2011] [Indexed: 11/20/2022]
Abstract
FHL2 displays tumor promoting or tumor suppressing activities depending on the types of tumor cells. In this study, we demonstrated that FHL2 overexpression inhibits the proliferation of human HCC cells Hep3B through cell cycle regulation by decreasing cyclin D1 expression while increasing the expressions of p21 and p27. FHL2 overexpression also inhibits migration and invasion of Hep3B cells through the regulation of epithelial-mesenchymal transition. Surprisingly, we also demonstrated an antiapoptotic function for FHL2 overexpression with increased resistance to doxorubicin-induced apoptosis, which indicates the separation of anti-proliferative and anti-apoptotic role of FHL2. Taken together, our results indicate FHL2 could exert anti-apoptotic effect independent of tumor growth suppression.
Collapse
|
73
|
Jin J, Kwon YW, Paek JS, Cho HJ, Yu J, Lee JY, Chu IS, Park IH, Park YB, Kim HS, Kim Y. Analysis of differential proteomes of induced pluripotent stem cells by protein-based reprogramming of fibroblasts. J Proteome Res 2011; 10:977-89. [PMID: 21175196 DOI: 10.1021/pr100624f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The recent generation of induced pluripotent stem (iPS) cells represents a novel opportunity to complement embryonic stem (ES) cell-based approaches. iPS cells can be generated by viral transduction of specific transcription factors, but there is a potential risk of tumorigenicity by random retroviral integration. We have generated novel iPS (sFB-protein-iPS) cells from murine dermal fibroblasts (FVB-sFB) that have ES cell characteristics, using ES cell-derived cell extracts instead of performing viral transduction. Notably, only cell extracts from an ES cell line (C57-mES) on the C57/BL6 background generated iPS cells in our protocol-not an ES cell line (E14-mES) on the 129 background. Hypothesizing that determining the differences in these 2 mES cell lines will provide vital insight into the reprogramming machinery, we performed proteomic and global gene expression analysis by iTRAQ and mRNA microarray, respectively. We observed that pluripotent ES cells and ES cell extract-derived iPS cells had differential proteomes and global gene expression patterns. Notably, reprogramming-competent C57-mES cells highly expressed proteins that regulate protein synthesis and metabolism, compared with reprogramming-incompetent 129-mES cells, suggesting that there is a threshold that protein synthetic machinery must exceed to initiate reprogramming.
Collapse
Affiliation(s)
- Jonghwa Jin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Zhang W, Yang Y, Jiang B, Peng J, Tu S, Sardet C, Zhang Y, Pang R, Hung IF, Tan VPY, Lam CSC, Wang J, Wong BC. XIAP-associated factor 1 interacts with and attenuates the trans-activity of four and a Half LIM protein 2. Mol Carcinog 2010; 50:199-207. [PMID: 21104993 DOI: 10.1002/mc.20705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 09/14/2010] [Accepted: 10/18/2010] [Indexed: 11/11/2022]
|
75
|
Himeda CL, Ranish JA, Pearson RCM, Crossley M, Hauschka SD. KLF3 regulates muscle-specific gene expression and synergizes with serum response factor on KLF binding sites. Mol Cell Biol 2010; 30:3430-43. [PMID: 20404088 PMCID: PMC2897560 DOI: 10.1128/mcb.00302-10] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 04/10/2010] [Indexed: 12/29/2022] Open
Abstract
This study identifies KLF3 as a transcriptional regulator of muscle genes and reveals a novel synergistic interaction between KLF3 and serum response factor (SRF). Using quantitative proteomics, KLF3 was identified as one of several candidate factors that recognize the MPEX control element in the Muscle creatine kinase (MCK) promoter. Chromatin immunoprecipitation analysis indicated that KLF3 is enriched at many muscle gene promoters (MCK, Myosin heavy chain IIa, Six4, Calcium channel receptor alpha-1, and Skeletal alpha-actin), and two KLF3 isoforms are upregulated during muscle differentiation. KLF3 and SRF physically associate and synergize in transactivating the MCK promoter independently of SRF binding to CArG motifs. The zinc finger and repression domains of KLF3 plus the MADS box and transcription activation domain of SRF are implicated in this synergy. Our results provide the first evidence of a role for KLF3 in muscle gene regulation and reveal an alternate mechanism for transcriptional regulation by SRF via its recruitment to KLF binding sites. Since both factors are expressed in all muscle lineages, SRF may regulate many striated- and smooth-muscle genes that lack known SRF control elements, thus further expanding the breadth of the emerging CArGome.
Collapse
Affiliation(s)
- Charis L. Himeda
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, Institute for Systems Biology, Seattle, Washington 98103-8904, School of Molecular and Microbial Biosciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Jeffrey A. Ranish
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, Institute for Systems Biology, Seattle, Washington 98103-8904, School of Molecular and Microbial Biosciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richard C. M. Pearson
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, Institute for Systems Biology, Seattle, Washington 98103-8904, School of Molecular and Microbial Biosciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Merlin Crossley
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, Institute for Systems Biology, Seattle, Washington 98103-8904, School of Molecular and Microbial Biosciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Stephen D. Hauschka
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, Institute for Systems Biology, Seattle, Washington 98103-8904, School of Molecular and Microbial Biosciences, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
76
|
Shi X, Bowlin KM, Garry DJ. Fhl2 interacts with Foxk1 and corepresses Foxo4 activity in myogenic progenitors. Stem Cells 2010; 28:462-9. [PMID: 20013826 DOI: 10.1002/stem.274] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adult skeletal muscle has a remarkable regenerative capacity because of a myogenic progenitor cell population. Using a gene disruption strategy, we determined that Foxk1 regulates myogenic progenitor cell activation and muscle regeneration. In this study, we undertook a yeast two hybrid screen to identify Foxk1 interacting proteins. We identified the LIM-only protein, Fhl2, as a Foxk1 interacting protein. Using transcriptional assays, we observed that Fhl2, in a dose-dependent fashion, promotes Foxk1 transcriptional repression of Foxo4 activity. Using histochemical and immunohistochemical assays, we further established that Fhl2 is expressed in the myogenic progenitor cell population. Fhl2 knockdown results in cell cycle arrest, and mice lacking Fhl2 have perturbed skeletal muscle regeneration. Collectively, these studies define a Fhl2-Foxk1 cascade that regulates the myogenic progenitor cell activity in adult skeletal muscle and enhances our understanding of muscle regeneration.
Collapse
Affiliation(s)
- Xiaozhong Shi
- Lillehei Heart Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
77
|
Pio R, Blanco D, Pajares MJ, Aibar E, Durany O, Ezponda T, Agorreta J, Gomez-Roman J, Anton MA, Rubio A, Lozano MD, López-Picazo JM, Subirada F, Maes T, Montuenga LM. Development of a novel splice array platform and its application in the identification of alternative splice variants in lung cancer. BMC Genomics 2010; 11:352. [PMID: 20525254 PMCID: PMC2889901 DOI: 10.1186/1471-2164-11-352] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 06/03/2010] [Indexed: 12/22/2022] Open
Abstract
Background Microarrays strategies, which allow for the characterization of thousands of alternative splice forms in a single test, can be applied to identify differential alternative splicing events. In this study, a novel splice array approach was developed, including the design of a high-density oligonucleotide array, a labeling procedure, and an algorithm to identify splice events. Results The array consisted of exon probes and thermodynamically balanced junction probes. Suboptimal probes were tagged and considered in the final analysis. An unbiased labeling protocol was developed using random primers. The algorithm used to distinguish changes in expression from changes in splicing was calibrated using internal non-spliced control sequences. The performance of this splice array was validated with artificial constructs for CDC6, VEGF, and PCBP4 isoforms. The platform was then applied to the analysis of differential splice forms in lung cancer samples compared to matched normal lung tissue. Overexpression of splice isoforms was identified for genes encoding CEACAM1, FHL-1, MLPH, and SUSD2. None of these splicing isoforms had been previously associated with lung cancer. Conclusions This methodology enables the detection of alternative splicing events in complex biological samples, providing a powerful tool to identify novel diagnostic and prognostic biomarkers for cancer and other pathologies.
Collapse
Affiliation(s)
- Ruben Pio
- Division of Oncology, Center for Applied Medical Research, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Wu H, Chen Y, Miao S, Zhang C, Zong S, Koide SS, Wang L. Sperm associated antigen 8 (SPAG8), a novel regulator of activator of CREM in testis during spermatogenesis. FEBS Lett 2010; 584:2807-15. [DOI: 10.1016/j.febslet.2010.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 05/04/2010] [Accepted: 05/08/2010] [Indexed: 10/19/2022]
|
79
|
Labalette C, Nouët Y, Levillayer F, Colnot S, Chen J, Claude V, Huerre M, Perret C, Buendia MA, Wei Y. Deficiency of the LIM-only protein FHL2 reduces intestinal tumorigenesis in Apc mutant mice. PLoS One 2010; 5:e10371. [PMID: 20442768 PMCID: PMC2860980 DOI: 10.1371/journal.pone.0010371] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 03/17/2010] [Indexed: 11/18/2022] Open
Abstract
Background The four and a half LIM-only protein 2 (FHL2) is capable of shuttling between focal adhesion and nucleus where it signals through direct interaction with a number of proteins including β-catenin. Although FHL2 activation has been found in various human cancers, evidence of its functional contribution to carcinogenesis has been lacking. Methodology/Principal Findings Here we have investigated the role of FHL2 in intestinal tumorigenesis in which activation of the Wnt pathway by mutations in the adenomatous polyposis coli gene (Apc) or in β-catenin constitutes the primary transforming event. In this murine model, introduction of a biallelic deletion of FHL2 into mutant ApcΔ14/+ mice substantially reduces the number of intestinal adenomas but not tumor growth, suggesting a role of FHL2 in the initial steps of tumorigenesis. In the lesions, Wnt signalling is not affected by FHL2 deficiency, remaining constitutively active. Nevertheless, loss of FHL2 activity is associated with increased epithelial cell migration in intestinal epithelium, which might allow to eliminate more efficiently deleterious cells and reduce the risk of tumorigenesis. This finding may provide a mechanistic basis for tumor suppression by FHL2 deficiency. In human colorectal carcinoma but not in low-grade dysplasia, we detected up-regulation and enhanced nuclear localization of FHL2, indicating the activation of FHL2 during the development of malignancy. Conclusions/Significance Our data demonstrate that FHL2 represents a critical factor in intestinal tumorigenesis.
Collapse
Affiliation(s)
- Charlotte Labalette
- Département de Virologie, Institut Pasteur, Paris, France
- Inserm U579, Paris, France
| | - Yann Nouët
- Département de Virologie, Institut Pasteur, Paris, France
- Inserm U579, Paris, France
| | - Florence Levillayer
- Département de Virologie, Institut Pasteur, Paris, France
- Inserm U579, Paris, France
| | - Sabine Colnot
- Département d'Endocrinologie Métabolisme et Cancer, Institut Cochin, Paris, France
- Inserm U567, Paris, France
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Valere Claude
- Département d'Anapathologie, Hôpital Bégin, Saint Mandé, France
| | - Michel Huerre
- Département d'Infection et Epidémiologie, Institut Pasteur, Paris, France
| | - Christine Perret
- Département d'Endocrinologie Métabolisme et Cancer, Institut Cochin, Paris, France
- Inserm U567, Paris, France
| | - Marie-Annick Buendia
- Département de Virologie, Institut Pasteur, Paris, France
- Inserm U579, Paris, France
| | - Yu Wei
- Département de Virologie, Institut Pasteur, Paris, France
- Inserm U579, Paris, France
- * E-mail:
| |
Collapse
|
80
|
Huang X, Wang JF, Xia WR, Zou MJ, Cai X, Xu DG. Identification of the transactivation domain of the human FHL3. Mol Biol 2010. [DOI: 10.1134/s0026893310020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
81
|
Cheung WKC, Yang PH, Huang QH, Chen Z, Chen SJ, Lin MCM, Kung HF. Identification of protein domains required for makorin-2-mediated neurogenesis inhibition in Xenopus embryos. Biochem Biophys Res Commun 2010; 394:18-23. [PMID: 20167204 DOI: 10.1016/j.bbrc.2010.02.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 02/08/2010] [Indexed: 11/15/2022]
Abstract
Makorin-2, consisting of four highly conserved C(3)H zinc fingers, a Cys-His motif and a C(3)HC(4) RING zinc finger domain, is a putative ribonucleoprotein. We have previously reported that Xenopus makorin-2 (mkrn2) is a neurogenesis inhibitor acting upstream of glycogen synthase kinase-3beta (GSK-3beta) in the phosphatidylinositol 3-kinase/Akt pathway. In an effort to identify the functional domains required for its anti-neurogenic activity, we designed and constructed a series of N- and C-terminal truncation mutants of mkrn2. Concurred with the full-length mkrn2, we showed that overexpression of one of the truncation mutants mkrn2(s)-7, which consists of only the third C(3)H zinc finger, Cys-His motif and C(3)HC(4) RING zinc finger, is essential and sufficient to produce the phenotypical dorso-posterior deficiencies and small-head/short-tail phenotype in tadpoles. In animal cap explant assay, we further demonstrated that mkrn2(s)-7 not only inhibits activin and retinoic acid-induced animal cap neuralization and the expression of a pan-neural marker neural cell adhesion molecule, but also induces GSK-3beta expression. These results collectively suggest that the third C(3)H zinc finger, Cys-His motif and C(3)HC(4) RING zinc finger are indispensable for the anti-neurogenic activity of mkrn2.
Collapse
Affiliation(s)
- William K C Cheung
- Department of Chemistry and Open Laboratory of Chemical Biology, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | | | | | | | | | | | | |
Collapse
|
82
|
|
83
|
Lardenois A, Chalmel F, Demougin P, Kotaja N, Sassone-Corsi P, Primig M. Fhl5/Act, a CREM-binding transcriptional activator required for normal sperm maturation and morphology, is not essential for testicular gene expression. Reprod Biol Endocrinol 2009; 7:133. [PMID: 19930692 PMCID: PMC2788571 DOI: 10.1186/1477-7827-7-133] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 11/24/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The LIM domain protein Fhl5 was previously found to interact with CREM, a DNA binding transcriptional regulator necessary for spermiogenesis in mammals. Co-transfection experiments using heterologous promoter constructs indicated a role for Fhl5 in transcriptional up-regulation of CREM-dependent testicular genes. Male mice lacking Fhl5 were reported to be fertile but displayed partially abnormal sperm maturation and morphology. METHODS To identify Fhl5 testicular target genes we carried out two whole-genome expression profiling experiments using high-density oligonucleotide microarrays and total testis samples from Fhl5 wild-type versus homozygous mutant mice first in different and then in isogenic strain backgrounds. RESULTS Weak signal differences were detected in non-isogenic samples but no statistically significant expression changes were observed when isogenic Fhl5 mutant and wild-type samples were compared. CONCLUSION The outcome of these experiments suggests that testicular expression profiling is extremely sensitive to the genetic background and that Fhl5 is not essential for testicular gene expression to a level detected by microarray-based measurements. This might be due to redundant function of the related and similarly expressed protein Fhl4.
Collapse
Affiliation(s)
| | - Frédéric Chalmel
- Inserm, U625, Université Rennes 1, IFR140, Rennes, F-35042, France
| | | | - Noora Kotaja
- University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| | | | - Michael Primig
- Inserm, U625, Université Rennes 1, IFR140, Rennes, F-35042, France
| |
Collapse
|
84
|
Ligand-specific sequential regulation of transcription factors for differentiation of MCF-7 cells. BMC Genomics 2009; 10:545. [PMID: 19925682 PMCID: PMC2785842 DOI: 10.1186/1471-2164-10-545] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 11/20/2009] [Indexed: 12/29/2022] Open
Abstract
Background Sharing a common ErbB/HER receptor signaling pathway, heregulin (HRG) induces differentiation of MCF-7 human breast cancer cells while epidermal growth factor (EGF) elicits proliferation. Although cell fates resulting from action of the aforementioned ligands completely different, the respective gene expression profiles in early transcription are qualitatively similar, suggesting that gene expression during late transcription, but not early transcription, may reflect ligand specificity. In this study, based on both the data from time-course quantitative real-time PCR on over 2,000 human transcription factors and microarray of all human genes, we identified a series of transcription factors which may control HRG-specific late transcription in MCF-7 cells. Results We predicted that four transcription factors including EGR4, FRA-1, FHL2, and DIPA should have responsibility of regulation in MCF-7 cell differentiation. Validation analysis suggested that one member of the activator protein 1 (AP-1) family, FOSL-1 (FRA-1 gene), appeared immediately following c-FOS expression, might be responsible for expression of transcription factor FHL2 through activation of the AP-1 complex. Furthermore, RNAi gene silencing of FOSL-1 and FHL2 resulted in increase of extracellular signal-regulated kinase (ERK) phosphorylation of which duration was sustained by HRG stimulation. Conclusion Our analysis indicated that a time-dependent transcriptional regulatory network including c-FOS, FRA-1, and FHL2 is vital in controlling the ERK signaling pathway through a negative feedback loop for MCF-7 cell differentiation.
Collapse
|
85
|
Cottle DL, McGrath MJ, Wilding BR, Cowling BS, Kane JM, D'Arcy CE, Holdsworth M, Hatzinisiriou I, Prescott M, Brown S, Mitchell CA. SLIMMER (FHL1B/KyoT3) interacts with the proapoptotic protein Siva-1 (CD27BP) and delays skeletal myoblast apoptosis. J Biol Chem 2009; 284:26964-77. [PMID: 19643733 DOI: 10.1074/jbc.m109.036293] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The fhl1 gene encoding four-and-a-half LIM protein-1 (FHL1) and its spliced isoform, SLIMMER, is mutated in reducing body myopathy, X-linked myopathy with postural muscle atrophy, scapuloperoneal myopathy, and rigid spine syndrome. In this study we have identified a novel function for SLIMMER in delaying skeletal muscle apoptosis via an interaction with the proapoptotic protein Siva-1. Siva-1 was identified as a SLIMMER-specific-interacting protein using yeast two-hybrid screening, direct-binding studies, and glutathione S-transferase pulldown analysis of murine skeletal muscle lysates. In C2C12 skeletal myoblasts, SLIMMER and Siva co-localized in the nucleus; however, both proteins exhibited redistribution to the cytoplasm following the differentiation of mononucleated myoblasts to multinucleated myotubes. In sections of mature skeletal muscle from wild type mice, SLIMMER and Siva-1 co-localized at the Z-line. SLIMMER and Siva-1 were also enriched in Pax-7-positive satellite cells, muscle stem cells that facilitate repair and regeneration. Significantly, SLIMMER delayed Siva-1-dependent apoptosis in C2C12 myoblasts. In skeletal muscle sections from the mdx mouse model of Duchenne muscular dystrophy, SLIMMER and Siva-1 co-localized in the nucleus of apoptotic myofibers. Therefore, SLIMMER may protect skeletal muscle from apoptosis.
Collapse
Affiliation(s)
- Denny L Cottle
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800 Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Schotman H, Karhinen L, Rabouille C. Integrins mediate their unconventional, mechanical-stress-induced secretion via RhoA and PINCH in Drosophila. J Cell Sci 2009; 122:2662-72. [PMID: 19584096 DOI: 10.1242/jcs.039347] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During the epithelium remodelling such as the flattening of the Drosophila follicular epithelium, the alpha-integrin subunits are unconventionally secreted through a dGRASP-dependent route that is built de novo. The biogenetic process starts with the upregulation of a small subset of targeted mRNAs, including dgrasp. Here, we show that dgrasp mRNA upregulation is triggered by the tension of the underlying oocyte and by applied external forces at the basal side of the follicular epithelium. We show that integrins are also involved in dgrasp mRNA upregulation and the epithelium remodelling. Tension leads to the recruitment of RhoA to the plasma membrane, where it participates in its remodelling. The LIM protein PINCH can cycle to the nucleus and is involved in dgrasp mRNA upregulation. We propose that integrins are involved in triggering the biogenesis of their own unconventional secretion route that they use to strengthen adhesion and ensure epithelial integrity at the next stages of development, perhaps by acting as mechanosensors of the underlying tension through RhoA and PINCH.
Collapse
Affiliation(s)
- Hans Schotman
- The Cell Microscopy Centre, Department of Cell Biology and Institute of Biomembrane, University Medical Centre Utrecht, AZU Rm G02.525, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | | | | |
Collapse
|
87
|
Manna PR, Dyson MT, Stocco DM. Role of basic leucine zipper proteins in transcriptional regulation of the steroidogenic acute regulatory protein gene. Mol Cell Endocrinol 2009; 302:1-11. [PMID: 19150388 PMCID: PMC5006949 DOI: 10.1016/j.mce.2008.12.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/15/2008] [Accepted: 12/15/2008] [Indexed: 01/23/2023]
Abstract
The regulation of steroidogenic acute regulatory protein (StAR) gene transcription by cAMP-dependent mechanisms occurs in the absence of a consensus cAMP response element (CRE, TGACGTGA). This regulation is coordinated by multiple transcription factors that bind to sequence-specific elements located approximately 150 bp upstream of the transcription start site. Among the proteins that bind within this region, the basic leucine zipper (bZIP) family of transcription factors, i.e. CRE binding protein (CREB)/CRE modulator (CREM)/activating transcription factor (ATF), activator protein 1 (AP-1; Fos/Jun), and CCAAT enhancer binding protein beta (C/EBPbeta), interact with an overlapping region (-81/-72 bp) in the StAR promoter, mediate stimulus-transcription coupling of cAMP signaling and play integral roles in regulating StAR gene expression. These bZIP proteins are structurally similar and bind to DNA sequences as dimers; however, they exhibit discrete transcriptional activities, interact with several transcription factors and other properties that contribute in their regulatory functions. The 5'-flanking -81/-72 bp region of the StAR gene appears to function as a key element within a complex cAMP response unit by binding to different bZIP members, and the StAR promoter displays variable states of cAMP responsivity contingent upon the occupancy of these cis-elements with these transcription factors. The expression and activities of CREB/CREM/ATF, Fos/Jun and C/EBPbeta have been demonstrated to be mediated by a plethora of extracellular signals, and the phosphorylation of these proteins at several Ser and Thr residues allows recruitment of the transcriptional coactivator CREB binding protein (CBP) or its functional homolog p300 to the StAR promoter. This review will focus on the current level of understanding of the roles of selective bZIP family proteins within the complex series of processes involved in regulating StAR gene transcription.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | |
Collapse
|
88
|
Hayashi H, Nakagami H, Takami Y, Koriyama H, Mori M, Tamai K, Sun J, Nagao K, Morishita R, Kaneda Y. FHL-2 suppresses VEGF-induced phosphatidylinositol 3-kinase/Akt activation via interaction with sphingosine kinase-1. Arterioscler Thromb Vasc Biol 2009; 29:909-14. [PMID: 19325137 DOI: 10.1161/atvbaha.108.178541] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE In the functional screening of a human heart cDNA library to identify a novel antiangiogenic factor, the prime candidate gene was "four-and-a-half LIM only protein-2" (FHL-2). The goal of this study is to clear the mechanism of antiangiogenic signaling of FHL-2 in endothelial cells (ECs). METHODS AND RESULTS Overexpressed FHL-2 strongly inhibited vascular endothelial growth factor (VEGF)-induced EC migration. In the angiogenic signaling, we focused on sphingosine kinase-1 (SK1), which produces sphingosine-1-phosphate (S1P), a bioactive sphingolipid, as a potent angiogenic mediator in ECs. Immunoprecipitation and immunostaining analysis showed that FHL-2 might bind to SK1. Importantly, overexpression of FHL-2 in ECs inhibited VEGF-induced SK1 activity, phosphatidylinositol 3-kinase activity, and phosphorylation of Akt and eNOS. In contrast, overexpression of FHL-2 had no effect on S1P-induced Akt phosphorylation. Interestingly, VEGF stimulation decreased the binding of FHL-2 and SK1. Depletion of FHL-2 by siRNA increased EC migration accompanied with SK1 and Akt activation, and increased the expression of VEGF receptor-2 which further enhanced VEGF signaling. Furthermore, injection of FHL-2 mRNA into Xenopus embryos resulted in inhibition of vascular network development, assessed by in situ hybridization with endothelial markers. CONCLUSIONS FHL-2 may regulate phosphatidylinositol 3-kinase/Akt via direct suppression of the SK1-S1P pathway in ECs.
Collapse
Affiliation(s)
- Hiroki Hayashi
- Division of Gene Therapy Science, Department of Geriatric Medicine, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita 565-0871, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Li M, Wang J, Ng SSM, Chan CY, Chen AC, Xia HP, Yew DT, Wong BCY, Chen Z, Kung HF, Lin MCM. The four-and-a-half-LIM protein 2 (FHL2) is overexpressed in gliomas and associated with oncogenic activities. Glia 2008; 56:1328-38. [PMID: 18615633 DOI: 10.1002/glia.20701] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Four-and-a-half-LIM protein 2 (FHL2) is a member of FHL protein family, which plays a crucial role in regulating gene expression, cell survival, and migration. Although its function in oncogenesis appears to be tumor type-specific, its roles in glioma formation and development are yet to be elucidated. In the present study, we demonstrated that the mRNA level of FHL2 was elevated in both low- and high-grade glioma samples. Overexpression of FHL2 stimulated the proliferation, anchorage-independent growth, and migration of human glioblastoma cells. Conversely, FHL2 knockdown by short hairpin RNA (shRNA-FHL2) inhibited glioblastoma cell proliferation and migration. Overexpression of FHL2 increased the tumorigenicity of glioblastoma cells in nude mice and decreased the mRNA levels of p53 and its downstream proapoptotic genes, including p21, Bcl2-associated protein X (Bax), and p53-upregulated modulator of apoptosis. It also enhanced the promoter activities of activator protein-1 (AP-1), human telomerase reverse transcriptase, and survivin genes. Together, these results provide the first evidence that FHL2 contributes to glioma carcinogenesis.
Collapse
Affiliation(s)
- Ming Li
- Department of Chemistry, Open laboratory of Chemical Biology, University of Hong Kong, Hong Kong, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
KyoT3, an isoform of murine FHL1, associates with the transcription factor RBP-J and represses the RBP-J-mediated transactivation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1779:805-10. [DOI: 10.1016/j.bbagrm.2008.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2007] [Revised: 07/31/2008] [Accepted: 08/01/2008] [Indexed: 11/23/2022]
|
91
|
The LIM-only protein FHL2 mediates ras-induced transformation through cyclin D1 and p53 pathways. PLoS One 2008; 3:e3761. [PMID: 19018287 PMCID: PMC2583050 DOI: 10.1371/journal.pone.0003761] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Accepted: 11/03/2008] [Indexed: 01/26/2023] Open
Abstract
Background Four and a half LIM-only protein 2 (FHL2) has been implicated in multiple signaling pathways that regulate cell growth and tissue homeostasis. We reported previously that FHL2 regulates cyclin D1 expression and that immortalized FHL2-null mouse embryo fibroblasts (MEFs) display reduced levels of cyclin D1 and low proliferative activity. Methodology/Principal Findings Here we address the contribution of FHL2 in cell transformation by investigating the effects of oncogenic Ras in FHL2-null context. We show that H-RasV12 provokes cell cycle arrest accompanied by accumulation of p53 and p16INK4a in immortalized FHL2−/− MEFs. These features contrast sharply with Ras transforming activity in wild type cell lines. We further show that establishment of FHL2-null cell lines differs from conventional immortalization scheme by retaining functional p19ARF/p53 checkpoint that is required for cell cycle arrest imposed by Ras. However, after serial passages of Ras-expressing FHL2−/− cells, dramatic increase in the levels of D-type cyclins and Rb phosphorylation correlates with the onset of cell proliferation and transformation without disrupting the p19ARF/p53 pathway. Interestingly, primary FHL2-null cells overexpressing cyclin D1 undergo a classical immortalization process leading to loss of the p19ARF/p53 checkpoint and susceptibility to Ras transformation. Conclusions/Significance Our findings uncover a novel aspect of cellular responses to mitogenic stimulation and illustrate a critical role of FHL2 in the signalling network that implicates Ras, cyclin D1 and p53.
Collapse
|
92
|
Qiao L, Wang Y, Pang R, Wang J, Dai Y, Ma J, Gu Q, Li Z, Zhang Y, Zou B, Lan HY, Wong BCY. Oncogene functions of FHL2 are independent from NF-kappaBIalpha in gastrointestinal cancer. Pathol Oncol Res 2008; 15:31-6. [PMID: 18752053 DOI: 10.1007/s12253-008-9085-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 07/07/2008] [Indexed: 11/30/2022]
Abstract
Four and a half of LIM-only protein 2 (FHL2) is an adaptor protein that can interact with many transcription factors and thus plays a variety of biological functions. Previous studies by our group have demonstrated that suppression of FHL2 was capable of inducing tumor cell differentiation, and inhibiting the growth of experimental gastric and colon cancers. Therefore, FHL2 appears to function as an oncogene. In order to further explore the mechanisms of how FHL2 is involved in tumorigenesis, we attempted to test whether FHL2 has any direct association with nuclear factor (NF-kappaB), the most important transcription factor involved in apoptosis, inflammation, and carcinogenesis. Using an Yeast Two Hybrid (Y2H) screening system, we have shown that FHL2 may have an interaction with NF-kappaBIalpha, the coding gene for IkappaBalpha which is the most potent endogenous inhibitor for NF-kappaB activation. However, subsequent studies using co-immunoprecipitation and co-localization failed to confirm the Y2H finding. Down-regulation of FHL2 by FHL2-siRNA down-regulated the expression of NF-kappaB p65. We therefore concluded that under the physiological condition, FHL2 may activate NF-kappaB pathway, even though such an activation may not be mediated by a direct binding of FHL2 to NF-kappaB inhibitor protein IkappaB.
Collapse
Affiliation(s)
- Liang Qiao
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Li X, Jia Z, Shen Y, Ichikawa H, Jarvik J, Nagele RG, Goldberg GS. Coordinate suppression of Sdpr and Fhl1 expression in tumors of the breast, kidney, and prostate. Cancer Sci 2008; 99:1326-33. [PMID: 18422756 PMCID: PMC11158056 DOI: 10.1111/j.1349-7006.2008.00816.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Src tyrosine kinase associates with the focal adhesion adaptor protein Cas (Crk-associated substrate) to suppress the expression of potential tumor suppressor genes. For example, Src utilizes Cas to suppress the expression of the LIM-only protein Fhl1 (four and a half LIM domains 1), in order to promote non-anchored tumor-cell growth and migration. Here, we report that the promoter region of the Fhl1 gene was methylated more in Src-transformed cells than non-transformed cells. In addition, global expression analysis indicates that Fhl1 induced expression of serum deprivation response factor (Sdpr) in Src-transformed cells. Moreover, Fhl1 and Sdpr was expressed in approximately 87% and 40% of samples obtained from non-transformed breast, 100% of samples obtained from non-transformed kidney, and over 60% of samples obtained from non-transformed prostate. In contrast, Fhl1 and Sdpr was detected in approximately 40% and 7% of matched samples from mammary carcinoma, less than 11% of matched samples from kidney carcinoma, and in less than 22% of matched samples from prostate carcinoma. These data indicate that Fhl1 and Sdpr expression was significantly reduced in tumors of the breast (P < 0.02 and P < 0.001), kidney (P < 0.01), and prostate (P < 0.05). In addition, although Src can activate mitogen-activated protein kinase (MAPK) to promote tumor-cell growth, our data indicate that Src did not rely on MAPK activity to suppress the expression of Fhl1 and Sdpr in transformed cells. Thus, Src induced methylation of the promoter region of the Fhl1 gene; Src suppressed Fhl1 and Sdpr expression independent of mitogen-activated protein kinase (MAPK) activity; Fhl1 induced the expression of Sdpr in Src-transformed cells; and Fhl1 and Sdpr expression was suppressed in tumors of the breast, kidney, and prostate.
Collapse
Affiliation(s)
- Xun Li
- Molecular Biology Department, University of Medicine and Dentistry of New Jersey, Stratford, NJ 08084, USA
| | | | | | | | | | | | | |
Collapse
|
94
|
Hinson JS, Medlin MD, Taylor JM, Mack CP. Regulation of myocardin factor protein stability by the LIM-only protein FHL2. Am J Physiol Heart Circ Physiol 2008; 295:H1067-H1075. [PMID: 18586895 DOI: 10.1152/ajpheart.91421.2007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Extensive evidence indicates that serum response factor (SRF) regulates muscle-specific gene expression and that myocardin family SRF cofactors are critical for smooth muscle cell differentiation. In a yeast two hybrid screen for novel SRF binding partners expressed in aortic SMC, we identified four and a half LIM domain protein 2 (FHL2) and confirmed this interaction by GST pull-down and coimmunoprecipitation assays. FHL2 also interacted with all three myocardin factors and enhanced myocardin and myocardin-related transcription factor (MRTF)-A-dependent transactivation of smooth muscle alpha-actin, SM22, and cardiac atrial natriuretic factor promoters in 10T1/2 cells. The expression of FHL2 increased myocardin and MRTF-A protein levels, and, importantly, this effect was due to an increase in protein stability not due to an increase in myocardin factor mRNA expression. Treatment of cells with proteasome inhibitors MG-132 and lactacystin strongly upregulated endogenous MRTF-A protein levels and resulted in a substantial increase in ubiquitin immunoreactivity in MRTF-A immunoprecipitants. Interestingly, the expression of FHL2 attenuated the effects of RhoA and MRTF-B on promoter activity, perhaps through decreased MRTF-B nuclear localization or decreased SRF-CArG binding. Taken together, these data indicate that myocardin factors are regulated by proteasome-mediated degradation and that FHL2 regulates SRF-dependent transcription by multiple mechanisms, including stabilization of myocardin and MRTF-A.
Collapse
Affiliation(s)
- Jeremiah S Hinson
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599-7525, USA
| | | | | | | |
Collapse
|
95
|
Labalette C, Nouët Y, Sobczak-Thepot J, Armengol C, Levillayer F, Gendron MC, Renard CA, Regnault B, Chen J, Buendia MA, Wei Y. The LIM-only protein FHL2 regulates cyclin D1 expression and cell proliferation. J Biol Chem 2008; 283:15201-8. [PMID: 18378678 DOI: 10.1074/jbc.m800708200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The LIM-only protein FHL2 acts as a transcriptional modulator that positively or negatively regulates multiple signaling pathways. We recently reported that FHL2 cooperates with CREB-binding protein/p300 in the activation of beta-catenin/T cell factor target gene cyclin D1. In this paper, we demonstrate that FHL2 is associated with the cyclin D1 promoter at the T cell factor/CRE site, providing evidence that cyclin D1 is a direct target of FHL2. We show that deficiency of FHL2 greatly reduces the proliferative capacity of spontaneously immortalized mouse fibroblasts, which is associated with decreased expression of cyclin D1 and p16(INK4a), and hypophosphorylation of Rb. Reexpression of FHL2 in FHL2-null fibroblasts efficiently restores cyclin D1 levels and cell proliferative capacity, indicating that FHL2 is critical for cyclin D1 activation and cell growth. Moreover, ectopic cyclin D1 expression is sufficient to override growth inhibition of immortalized FHL2-null fibroblasts. Gene expression profiling revealed that FHL2 deficiency triggers a broad change of the cell cycle program that is associated with down-regulation of several G(1)/S and G(2)/M cyclins, E2F transcription factors, and DNA replication machinery, thus correlating with reduced cell proliferation. This change also involves down-regulation of the negative cell cycle regulators, particularly INK4 inhibitors, which could counteract the decreased expression of cyclins, allowing cells to grow. Our study illustrates that FHL2 can act on different aspects of the cell cycle program to finely regulate cell proliferation.
Collapse
Affiliation(s)
- Charlotte Labalette
- Unité d'Oncogenèse et Virologie Moléculaire and PT Puce à ADN, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
Expression of Ldhc begins with the onset of meiosis in male germ cells and continues throughout spermatogenesis. Transcriptional regulatory mechanisms, especially in primary spermatocytes, are poorly described because of the lack of a reliable cell culture system. We constructed mouse transgenics and transfected germ cells in situ to study expression of the testis-specific isozyme of lactate dehydrogenase (LDH). From previous work, we determined that a 100-bp Ldhc core promoter contained potential cis regulatory elements, including a palindrome (-21 to +10), GC box (-70 to -65), and cAMP-responsive element (CRE) sites (-53 to -49, -39 to -35). We provide here the demonstration of a functional role for these sequences by expression of mutated transgenes in vivo. Our results reveal for the first time that mutation of the GC box does not abolish promoter activity, which remains testis-specific. Mutation of GC box or CRE sites resulted in a 73% and 74% reduction in promoter activity, respectively, in a transient transfection of germ cells in vivo by electroporation; the combination of GC box and CRE site mutations eliminates promoter activity. Therefore, we conclude that simultaneous occupancy of the GC box and CRE sites in the core promoter is necessary for full expression of Ldhc in the testis.
Collapse
Affiliation(s)
- HuangHui Tang
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, Illinois 60208
| | - Aisha Kung
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, Illinois 60208
| | - Erwin Goldberg
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
97
|
Wang LL, Fu WN, Li ZG, Sun KL. [Research of HOXD13 and FHL1 in idiopathic congenital talipes equinovarus]. YI CHUAN = HEREDITAS 2008; 30:46-50. [PMID: 18244901 DOI: 10.3724/sp.j.1005.2008.00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To investigate the relationship of HOXD13 and FHL1 in idiopathic congenital talipes equinovarus(ICTEV), 84 samples from patients with ICTEV were used in the study. Mutation in the coding region of HOXD13 was detected by denaturing gradinent electrophoresis. The mRNA and protein levels of HOXD13 and FHL1 were evaluated by RT-PCR and immunohistochemistry, respectively. The binding site of FHL1 to HOXD13 predicted by PMATCH software was validated by EMSA( Electrophoretic mobility shift assay,EMSA).No mutation was found in the coding region of HOXD13 in 84 samples from patients with ICTEV. Both HOXD13(33.3%) and FHL1(46.6%) were down-regulated in ICTEV muscle tissue. The result of EMSA showed that the special binding band appeared when HOXD13 existed. The results shows that HOXD13 gene mutation was not involved in outbreak in idiopathic congenital talipes equinovarus, but changes of HOXD13 and FHL1 gene expression related to the development of talipes equinovarus malformation. HOXD13 might play an role in ICTEV through regulating FHL1 expression.
Collapse
Affiliation(s)
- Li-Li Wang
- Department of Medical Genetics, China Medical University, Shenyang, 110001, China.
| | | | | | | |
Collapse
|
98
|
Protein kinase A, Ca2+/calmodulin-dependent kinase II, and calcineurin regulate the intracellular trafficking of myopodin between the Z-disc and the nucleus of cardiac myocytes. Mol Cell Biol 2007; 27:8215-27. [PMID: 17923693 DOI: 10.1128/mcb.00950-07] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Spatial and temporal resolution of intracellular signaling can be achieved by compartmentalizing transduction units. Myopodin is a dual-compartment, actin-bundling protein that shuttles between the nucleus and the Z-disc of myocytes in a differentiation- and stress-dependent fashion. Importin alpha binding and nuclear import of myopodin are regulated by serine/threonine phosphorylation-dependent binding of myopodin to 14-3-3. Here we show that in the heart myopodin forms a Z-disc signaling complex with alpha-actinin, calcineurin, Ca2+/calmodulin-dependent kinase II (CaMKII), muscle-specific A-kinase anchoring protein, and myomegalin. Phosphorylation of myopodin by protein kinase A (PKA) or CaMKII mediates 14-3-3 binding and nuclear import in myoblasts. Dephosphorylation of myopodin by calcineurin abrogates 14-3-3beta binding. Activation of PKA or inhibition of calcineurin in adult cardiac myocytes releases myopodin from the Z-disc and induces its nuclear import. The identification of myopodin as a direct target of PKA, CaMKII, and calcineurin defines a novel intracellular signaling pathway whereby changes in Z-disc dynamics may translate into compartmentalized signal transduction in the heart.
Collapse
|
99
|
Takemori H, Kajimura J, Okamoto M. TORC-SIK cascade regulates CREB activity through the basic leucine zipper domain. FEBS J 2007; 274:3202-9. [PMID: 17565599 DOI: 10.1111/j.1742-4658.2007.05889.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The transcription factor cAMP response element-binding protein (CREB) plays important roles in gene expression induced by cAMP signaling and is believed to be activated when its Ser133 is phosphorylated. However, the discovery of Ser133-independent activation by the activation of transducer of regulated CREB activity coactivators (TORC) and repression by salt inducible kinase cascades suggests that Ser133-independent regulation of CREB is also important. The activation and repression are mediated by the basic leucine zipper domain of CREB. In this review, we focus on the basic leucine zipper domain in the regulation of transcriptional activity of CREB and describe the functions of TORC and salt inducible kinase.
Collapse
Affiliation(s)
- Hiroshi Takemori
- Laboratory of Cell Signaling and Metabolism, National Institute of Biomedical Innovation, Ibaraki, Osaka, Japan.
| | | | | |
Collapse
|
100
|
Skopeliti M, Kratzer U, Altenberend F, Panayotou G, Kalbacher H, Stevanovic S, Voelter W, Tsitsilonis OE. Proteomic exploitation on prothymosin α-induced mononuclear cell activation. Proteomics 2007; 7:1814-24. [PMID: 17474146 DOI: 10.1002/pmic.200600870] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Prothymosin alpha (ProTalpha) is an acidic polypeptide associated both with cell proliferation and immune regulation. Although ProTalpha's immunomodulating activity is well established at cellular level, limited information is available regarding the signaling pathways triggered by ProTalpha. Using 2-DE proteomic technology, we investigated changes in protein expression of ProTalpha-stimulated peripheral blood mononuclear cells (PBMC) in the course of a 3-day incubation. Using healthy donor- and cancer patient-derived PBMC, 12 gels were studied, identifying 53 differing protein spots via PMF comparison analysis. Among others, we identified interleukin-1 receptor-associated kinase 4, heat-shock protein 90, lipocalin 2, ribophorin 1, eukaryotic elongation factor 2, 14-3-3 protein, L-plastin, and MX2 protein, all of which were found to be overexpressed upon ProTalpha activation. Based on the physiological role of upregulated proteins, we propose the following model for ProTalpha's immunological mode of action: on day 1, ProTalpha triggers monocyte activation, possibly via toll-like receptor signaling, and enhances antigen presentation, consequently promoting and stabilizing monocyte-T-cell immune synapse; on day 2, activated monocytes produce interleukin (IL)-1, while T-cell receptor triggering promotes T-cell proliferation and IL-2 production; finally, on day 3, ProTalpha-activated PBMC express proteins related to adhesion and cytotoxic effector functions, both contributing to the increase of their lytic activity.
Collapse
Affiliation(s)
- Margarita Skopeliti
- Department of Animal and Human Physiology, Faculty of Biology, University of Athens, Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|