1
|
Hermant C, Mourra-Díaz CM, Oomen ME, Altamirano-Pacheco L, Pal M, Nakatani T, Torres-Padilla ME. The transcription factor SRF regulates MERVL retrotransposons and gene expression during zygotic genome activation. Genes Dev 2025; 39:490-509. [PMID: 40015990 PMCID: PMC11960700 DOI: 10.1101/gad.352270.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025]
Abstract
The regulatory circuitry of cell-specific transcriptional programs is thought to be influenced by transposable elements (TEs), whereby TEs serve as raw material for the diversification and genome-wide distribution of genetic elements that contain cis-regulatory activity. However, the transcriptional activators of TEs in relevant physiological contexts are largely unknown. Here, we undertook an evolutionary approach to identify regulators of two main families of MERVL, a major regulator of transcription during early mouse development. Using a combination of phyloregulatory, transcriptomic, and loss-of-function approaches, we demonstrate that SRF is a novel regulator of MERVL and embryonic transcription during zygotic genome activation. By resolving the phylogenetic history of two major MERVL families, we delineate the evolutionary acquisition of SRF and DUX binding sites and show that the acquisition of the SRF site precedes that of DUX. SRF contributes to embryonic transcription through the regulation of MERVLs, which in turn serve as promoters for host genes. Our work identifies new transcriptional regulators and TEs that shape the gene expression programs in early embryos and highlights the process of TE domestication via the sequential acquisition of transcription factor binding sites and coevolution with the host.
Collapse
Affiliation(s)
- Clara Hermant
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany
| | | | - Marlies E Oomen
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany
| | - Luis Altamirano-Pacheco
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany
| | - Mrinmoy Pal
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany
| | - Tsunetoshi Nakatani
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany
| | - Maria-Elena Torres-Padilla
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377 München, Germany;
- Faculty of Biology, Ludwig-Maximilians Universität, D-81377 München, Germany
| |
Collapse
|
2
|
Deshpande A, Shetty PMV, Frey N, Rangrez AY. SRF: a seriously responsible factor in cardiac development and disease. J Biomed Sci 2022; 29:38. [PMID: 35681202 PMCID: PMC9185982 DOI: 10.1186/s12929-022-00820-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
The molecular mechanisms that regulate embryogenesis and cardiac development are calibrated by multiple signal transduction pathways within or between different cell lineages via autocrine or paracrine mechanisms of action. The heart is the first functional organ to form during development, which highlights the importance of this organ in later stages of growth. Knowledge of the regulatory mechanisms underlying cardiac development and adult cardiac homeostasis paves the way for discovering therapeutic possibilities for cardiac disease treatment. Serum response factor (SRF) is a major transcription factor that controls both embryonic and adult cardiac development. SRF expression is needed through the duration of development, from the first mesodermal cell in a developing embryo to the last cell damaged by infarction in the myocardium. Precise regulation of SRF expression is critical for mesoderm formation and cardiac crescent formation in the embryo, and altered SRF levels lead to cardiomyopathies in the adult heart, suggesting the vital role played by SRF in cardiac development and disease. This review provides a detailed overview of SRF and its partners in their various functions and discusses the future scope and possible therapeutic potential of SRF in the cardiovascular system.
Collapse
Affiliation(s)
- Anushka Deshpande
- Department of Internal Medicine III, Cardiology and Angiology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Prithviraj Manohar Vijaya Shetty
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
3
|
Azam H, Pierro L, Reina M, Gallagher WM, Prencipe M. Emerging role for the Serum Response Factor (SRF) as a potential therapeutic target in cancer. Expert Opin Ther Targets 2022; 26:155-169. [PMID: 35114091 DOI: 10.1080/14728222.2022.2032652] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The Serum Response Factor (SRF) is a transcription factor involved in three hallmarks of cancer: the promotion of cell proliferation, cell death resistance and invasion and metastasis induction. Many studies have demonstrated a leading role in the development and progression of multiple cancer types, thus highlighting the potential of SRF as a prognostic biomarker and therapeutic target, especially for cancers with poor prognosis. AREAS COVERED This review examines the role of SRF in several cancers in promoting cellular processes associated with cancer development and progression. SRF co-factors and signalling pathways are discussed as possible targets to inhibit SRF in a tissue and cancer-specific way. Small-molecule inhibitors of SRF, such as the CCGs series of compounds and lestaurtinib, which could be used as cancer therapeutics, are also discussed. EXPERT OPINION Targeting of SRF and its co-factors represents a promising therapeutic approach. Further understanding of the molecular mechanisms behind the action of SRF could provide a pipeline of novel molecular targets and therapeutic combinations for cancer. Basket clinical trials and the use of SRF immunohistochemistry as companion diagnostics will help testing of these new targets in patients.
Collapse
Affiliation(s)
- Haleema Azam
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Lisa Pierro
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Martina Reina
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - William M Gallagher
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Maria Prencipe
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| |
Collapse
|
4
|
Li R, Ong Q, Wong CC, Chu ESH, Sung JJY, Yang X, Yu J. O-GlcNAcylation inhibits hepatic stellate cell activation. J Gastroenterol Hepatol 2021; 36:3477-3486. [PMID: 34547820 PMCID: PMC11648924 DOI: 10.1111/jgh.15690] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/12/2021] [Accepted: 09/12/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND AIM Protein O-GlcNAcylation is a critical post-translational modification regulating gene expression and fundamental cell functions. O-GlcNAc transferase (OGT) emerged as a key regulator of liver pathophysiology and disease. In this study, we aimed to evaluate the role of OGT in hepatic stellate cells (HSCs) and its consequent role in liver fibrosis. METHODS Primary HSCs were isolated from C57/B6 mice. Cell morphology and α-SMA immunofluorescence staining were observed under scanning confocal microscope. Transcriptomic profile was evaluated by RNAseq (Illumina). Promoter activity was examined by luciferase and β--Galactosidase reporter assays. Liver fibrosis mouse models were induced either by intraperitoneal injection of CCl4 at 3 times/week for 4 weeks or by feeding with methionine and choline deficient (MCD) diet for 4 weeks. RESULTS OGT protein expression and protein O-GlcNAcylation were significantly decreased in CCl4 - or MCD diet-induced liver fibrosis as compared with normal liver in mice. OGT expression and protein O-GlcNAcylation were also decreased in primary HSCs isolated from liver with CCl4 -induced fibrosis compared with those from normal liver. RNA-seq showed that OGT knockdown in HSCs modulated key signaling pathways involved in HSC activation. Promoter sequence analysis of the differentially expressed genes predicted serum response factor (SRF) as a key transcription factor regulated by OGT. Luciferase reporter assay confirmed that OGT repressed activity of SRF to induce α-SMA transcription. Mutations of specific O-GlcNAcylation sites on SRF increased its transcriptional activity, validating negative regulation of SRF by OGT-mediated O-GlcNAcylation. CONCLUSIONS Our results suggest that OGT functions as a negative regulator of HSC activation by promoting SRF O-GlcNAcylation to protect against liver fibrosis.
Collapse
Affiliation(s)
- Rui Li
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Comparative Medicine, Department of Cellular and Molecular Physiology, Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Qunxiang Ong
- Department of Comparative Medicine, Department of Cellular and Molecular Physiology, Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chi Chun Wong
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eagle SH Chu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joseph JY Sung
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaoyong Yang
- Department of Comparative Medicine, Department of Cellular and Molecular Physiology, Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Woulfe KC, Jeffrey DA, Pires Da Silva J, Wilson CE, Mahaffey JH, Lau E, Slavov D, Hailu F, Karimpour-Fard A, Dockstader K, Bristow MR, Stauffer BL, Miyamoto SD, Sucharov CC. Serum response factor deletion 5 regulates phospholamban phosphorylation and calcium uptake. J Mol Cell Cardiol 2021; 159:28-37. [PMID: 34139234 PMCID: PMC8546760 DOI: 10.1016/j.yjmcc.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/25/2021] [Accepted: 06/13/2021] [Indexed: 11/25/2022]
Abstract
AIMS Pediatric dilated cardiomyopathy (pDCM) is characterized by unique age-dependent molecular mechanisms that include myocellular responses to therapy. We previously showed that pDCM, but not adult DCM patients respond to phosphodiesterase 3 inhibitors (PDE3i) by increasing levels of the second messenger cAMP and consequent phosphorylation of phospholamban (PLN). However, the molecular mechanisms involved in the differential pediatric and adult response to PDE3i are not clear. METHODS AND RESULTS Quantification of serum response factor (SRF) isoforms from the left ventricle of explanted hearts showed that PDE3i treatment affects expression of SRF isoforms in pDCM hearts. An SRF isoform lacking exon 5 (SRFdel5) was highly expressed in the hearts of pediatric, but not adult DCM patients treated with PDE3i. To determine the functional consequence of expression of SRFdel5, we overexpressed full length SRF or SRFdel5 in cultured cardiomyocytes with and without adrenergic stimulation. Compared to a control adenovirus, expression of SRFdel5 increased phosphorylation of PLN, negatively affected expression of the phosphatase that promotes dephosphorylation of PLN (PP2Cε), and promoted faster calcium reuptake, whereas expression of full length SRF attenuated calcium reuptake through blunted phosphorylation of PLN. CONCLUSIONS Taken together, these data indicate that expression of SRFdel5 in pDCM hearts in response to PDE3i contributes to improved function through regulating PLN phosphorylation and thereby calcium reuptake.
Collapse
Affiliation(s)
- Kathleen C Woulfe
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Danielle A Jeffrey
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Julie Pires Da Silva
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cortney E Wilson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer H Mahaffey
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Edward Lau
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dobromir Slavov
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Frehiwet Hailu
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Anis Karimpour-Fard
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karen Dockstader
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michael R Bristow
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Brian L Stauffer
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Denver Health Medical Center, Denver, CO, United States
| | - Shelley D Miyamoto
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital of Colorado, Aurora, CO, United States
| | - Carmen C Sucharov
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
6
|
Aberdeen H, Battles K, Taylor A, Garner-Donald J, Davis-Wilson A, Rogers BT, Cavalier C, Williams ED. The Aging Vasculature: Glucose Tolerance, Hypoglycemia and the Role of the Serum Response Factor. J Cardiovasc Dev Dis 2021; 8:58. [PMID: 34067715 PMCID: PMC8156687 DOI: 10.3390/jcdd8050058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
The fastest growing demographic in the U.S. at the present time is those aged 65 years and older. Accompanying advancing age are a myriad of physiological changes in which reserve capacity is diminished and homeostatic control attenuates. One facet of homeostatic control lost with advancing age is glucose tolerance. Nowhere is this more accentuated than in the high proportion of older Americans who are diabetic. Coupled with advancing age, diabetes predisposes affected subjects to the onset and progression of cardiovascular disease (CVD). In the treatment of type 2 diabetes, hypoglycemic episodes are a frequent clinical manifestation, which often result in more severe pathological outcomes compared to those observed in cases of insulin resistance, including premature appearance of biomarkers of senescence. Unfortunately, molecular mechanisms of hypoglycemia remain unclear and the subject of much debate. In this review, the molecular basis of the aging vasculature (endothelium) and how glycemic flux drives the appearance of cardiovascular lesions and injury are discussed. Further, we review the potential role of the serum response factor (SRF) in driving glycemic flux-related cellular signaling through its association with various proteins.
Collapse
Affiliation(s)
- Hazel Aberdeen
- Department of Biomedical Sciences, Baptist Health Sciences University, Memphis, TN 38103, USA; or
| | - Kaela Battles
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Ariana Taylor
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Jeranae Garner-Donald
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Ana Davis-Wilson
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Bryan T. Rogers
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Candice Cavalier
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| | - Emmanuel D. Williams
- Department of Biology and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA; (K.B.); (A.T.); (J.G.-D.); (A.D.-W.); (B.T.R.); (C.C.)
| |
Collapse
|
7
|
Caines R, Cochrane A, Kelaini S, Vila-Gonzalez M, Yang C, Eleftheriadou M, Moez A, Stitt AW, Zeng L, Grieve DJ, Margariti A. The RNA-binding protein QKI controls alternative splicing in vascular cells, producing an effective model for therapy. J Cell Sci 2019; 132:jcs.230276. [PMID: 31331967 DOI: 10.1242/jcs.230276] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/10/2019] [Indexed: 12/31/2022] Open
Abstract
Dysfunction of endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) leads to ischaemia, the central pathology of cardiovascular disease. Stem cell technology will revolutionise regenerative medicine, but a need remains to understand key mechanisms of vascular differentiation. RNA-binding proteins have emerged as novel post-transcriptional regulators of alternative splicing and we have previously shown that the RNA-binding protein Quaking (QKI) plays roles in EC differentiation. In this study, we decipher the role of the alternative splicing isoform Quaking 6 (QKI-6) to induce VSMC differentiation from induced pluripotent stem cells (iPSCs). PDGF-BB stimulation induced QKI-6, which bound to HDAC7 intron 1 via the QKI-binding motif, promoting HDAC7 splicing and iPS-VSMC differentiation. Overexpression of QKI-6 transcriptionally activated SM22 (also known as TAGLN), while QKI-6 knockdown diminished differentiation capability. VSMCs overexpressing QKI-6 demonstrated greater contractile ability, and upon combination with iPS-ECs-overexpressing the alternative splicing isoform Quaking 5 (QKI-5), exhibited higher angiogenic potential in vivo than control cells alone. This study demonstrates that QKI-6 is critical for modulation of HDAC7 splicing, regulating phenotypically and functionally robust iPS-VSMCs. These findings also highlight that the QKI isoforms hold key roles in alternative splicing, giving rise to cells which can be used in vascular therapy or for disease modelling.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Rachel Caines
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Amy Cochrane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Sophia Kelaini
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Marta Vila-Gonzalez
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Chunbo Yang
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Magdalini Eleftheriadou
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Arya Moez
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Lingfang Zeng
- Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - David J Grieve
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| | - Andriana Margariti
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL
| |
Collapse
|
8
|
Gerosa L, Grillo B, Forastieri C, Longaretti A, Toffolo E, Mallei A, Bassani S, Popoli M, Battaglioli E, Rusconi F. SRF and SRFΔ5 Splicing Isoform Recruit Corepressor LSD1/KDM1A Modifying Structural Neuroplasticity and Environmental Stress Response. Mol Neurobiol 2019; 57:393-407. [DOI: 10.1007/s12035-019-01720-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
Harada N, Okuyama M, Yoshikatsu A, Yamamoto H, Ishiwata S, Hamada C, Hirose T, Shono M, Kuroda M, Tsutsumi R, Takeo J, Taketani Y, Nakaya Y, Sakaue H. Endoplasmic Reticulum Stress in Mice Increases Hepatic Expression of Genes Carrying a Premature Termination Codon via a Nutritional Status‐Independent GRP78‐Dependent Mechanism. J Cell Biochem 2017; 118:3810-3824. [DOI: 10.1002/jcb.26031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/04/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Nagakatsu Harada
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Maiko Okuyama
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Aya Yoshikatsu
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Hironori Yamamoto
- Faculty of Human LifeDepartment of Health and NutritionJin‐ai University3‐1‐1 Ohde‐choEchizen City915‐8586Japan
| | - Saori Ishiwata
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Chikako Hamada
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Tomoyo Hirose
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Masayuki Shono
- Support Center for Advanced Medical SciencesInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Masashi Kuroda
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Rie Tsutsumi
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Jiro Takeo
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
- Central Research LaboratoryNippon Suisan Kaisha32‐3 Nanakuni 1 ChomeHachiojiTokyo192‐0991Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food ManagementInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Yutaka Nakaya
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| | - Hiroshi Sakaue
- Department of Nutrition and MetabolismInstitute of Biomedical SciencesTokushima University Graduate School3‐18‐15, Kuramoto‐choTokushima City770‐8503Japan
| |
Collapse
|
10
|
Meyer zu Reckendorf C, Anastasiadou S, Bachhuber F, Franz-Wachtel M, Macek B, Knöll B. Proteomic analysis of SRF associated transcription complexes identified TFII-I as modulator of SRF function in neurons. Eur J Cell Biol 2016; 95:42-56. [DOI: 10.1016/j.ejcb.2015.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 11/25/2022] Open
|
11
|
Likhoshvai VA, Kogai VV, Fadeev SI, Khlebodarova TM. Alternative splicing can lead to chaos. J Bioinform Comput Biol 2015; 13:1540003. [PMID: 25556917 DOI: 10.1142/s021972001540003x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Alternative splicing is a widespread phenomenon in higher eukaryotes, where it serves as a mechanism to increase the functional diversity of proteins. This phenomenon has been described for different classes of proteins, including transcription regulatory proteins. We demonstrated that in the simplest genetic system model the formation of the alternatively spliced isoforms with opposite functions (activators and repressors) could be a cause of transition to chaotic dynamics. Under the simplest genetic system we understand a system consisting of a single gene encoding the structure of a transcription regulatory protein whose expression is regulated by a feedback mechanism. As demonstrated by numerical analysis of the models, if the synthesized isoforms regulate the expression of their own gene acting through different sites and independently of each other, for the generation of chaotic dynamics it is sufficient that the regulatory proteins have a dimeric structure. If regulatory proteins act through one site, the chaotic dynamics is generated in the system only when the repressor protein is either a tetrameric or a higher-dimensional multimer. In this case the activator can be a dimer. It was also demonstrated that if the transcription factor isoforms exhibit either activating or inhibiting activity and are lower-dimensional multimers (< 4), independently of the regulation type the model demonstrates either cyclic or stationary trajectories.
Collapse
Affiliation(s)
- Vitaly A Likhoshvai
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, pr. Lavrentieva 10, Novosibirsk 630090, Russia , Novosibirsk State University, av. Pirogova 2, Novosibirsk 630090, Russia
| | | | | | | |
Collapse
|
12
|
Graeff M, Wenkel S. Regulation of protein function by interfering protein species. Biomol Concepts 2014; 3:71-8. [PMID: 25436525 DOI: 10.1515/bmc.2011.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 11/02/2011] [Indexed: 11/15/2022] Open
Abstract
Abstract Most proteins do not function alone but act in protein complexes. For several transcriptional regulators, it is known that they have to homo- or heterodimerize prior to DNA binding. These protein interactions occur through defined protein-protein-interaction (PPI) domains. More than two decades ago, inhibitor of DNA binding (ID), a small protein containing a single helix-loop-helix (HLH) motif was identified. ID is able to interact with the larger DNA-binding basic helix-loop-helix (bHLH) transcription factors, but due to the lack of the basic domain required for DNA binding, ID traps bHLH proteins in non-functional complexes. Work in plants has, in the recent years, identified more small proteins acting in analogy to ID. A hallmark of these small negative acting proteins is the presence of a protein-interaction domain and the absence of other functional domains required for transcriptional activation or DNA binding. Because these proteins are often very small and function in analogy to microRNAs (meaning in a dominant-negative manner), we propose to refer to these protein species as 'microProteins' (miPs). miPs can be encoded in the genome as individual transcription units but can also be produced by alternative splicing. Other negatively acting proteins, consisting of more than one domain, have also been identified, and we propose to call these proteins 'interfering proteins' (iPs). The aim of this review is to state more precisely how to discriminate miPs from iPs. Therefore, we will highlight recent findings on both protein species and describe their mode of action. Furthermore, miPs have the ability to regulate proteins of diverse functions, emphasizing their value as biotechnological tools.
Collapse
|
13
|
Vasudevan HN, Soriano P. SRF regulates craniofacial development through selective recruitment of MRTF cofactors by PDGF signaling. Dev Cell 2014; 31:332-344. [PMID: 25453829 DOI: 10.1016/j.devcel.2014.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/02/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023]
Abstract
Receptor tyrosine kinase signaling is critical for mammalian craniofacial development, but the key downstream transcriptional effectors remain unknown. We demonstrate that serum response factor (SRF) is induced by both platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) signaling in mouse embryonic palatal mesenchyme cells and that Srf neural crest conditional mutants exhibit facial clefting accompanied by proliferation and migration defects. Srf and Pdgfra mutants interact genetically in craniofacial development, but Srf and Fgfr1 mutants do not. This signal specificity is recapitulated at the level of cofactor activation: while both PDGF and FGF target gene promoters show enriched genome-wide overlap with SRF ChIP-seq peaks, PDGF selectively activates a network of MRTF-dependent cytoskeletal genes. Collectively, our results identify a role for SRF in proliferation and migration during craniofacial development and delineate a mechanism of receptor tyrosine kinase specificity mediated through differential cofactor usage, leading to a PDGF-responsive SRF-driven transcriptional program in the midface.
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
14
|
Glidewell-Kenney CA, Trang C, Shao PP, Gutierrez-Reed N, Uzo-Okereke AM, Coss D, Mellon PL. Neurokinin B induces c-fos transcription via protein kinase C and activation of serum response factor and Elk-1 in immortalized GnRH neurons. Endocrinology 2014; 155:3909-19. [PMID: 25057795 PMCID: PMC4164922 DOI: 10.1210/en.2014-1263] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mutations in neurokinin B (NKB) and its receptor, NK3R, were identified in human patients with hypogonadotropic hypogonadism, a disorder characterized by lack of puberty and infertility. Further studies have suggested that NKB acts at the level of the hypothalamus to control GnRH neuron activity, either directly or indirectly. We recently reported that treatment with senktide, a NK3R agonist, induced GnRH secretion and expression of c-fos mRNA in GT1-7 cells. Here, we map the responsive region in the murine c-fos promoter to between -400 and -200 bp, identify the signal transducer and activator of transcription (STAT) (-345) and serum response element (-310) sites as required for induction, a modulatory role for the Ets site (-318), and show that induction is protein kinase C dependent. Using gel shift and Gal4 assays, we further show that phosphorylation of Elk-1 leads to binding to DNA in complex with serum response factor at serum response element and Ets sites within the c-fos promoter. Thus, we determine molecular mechanisms involved in NKB regulation of c-fos induction, which may play a role in modulation of GnRH neuron activation.
Collapse
Affiliation(s)
- Christine A Glidewell-Kenney
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674
| | | | | | | | | | | | | |
Collapse
|
15
|
Blech-Hermoni Y, Ladd AN. RNA binding proteins in the regulation of heart development. Int J Biochem Cell Biol 2013; 45:2467-78. [PMID: 23973289 DOI: 10.1016/j.biocel.2013.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 11/28/2022]
Abstract
In vivo, RNA molecules are constantly accompanied by RNA binding proteins (RBPs), which are intimately involved in every step of RNA biology, including transcription, editing, splicing, transport and localization, stability, and translation. RBPs therefore have opportunities to shape gene expression at multiple levels. This capacity is particularly important during development, when dynamic chemical and physical changes give rise to complex organs and tissues. This review discusses RBPs in the context of heart development. Since the targets and functions of most RBPs--in the heart and at large--are not fully understood, this review focuses on the expression and roles of RBPs that have been implicated in specific stages of heart development or developmental pathology. RBPs are involved in nearly every stage of cardiogenesis, including the formation, morphogenesis, and maturation of the heart. A fuller understanding of the roles and substrates of these proteins could ultimately provide attractive targets for the design of therapies for congenital heart defects, cardiovascular disease, or cardiac tissue repair.
Collapse
Affiliation(s)
- Yotam Blech-Hermoni
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Program in Cell Biology, Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
16
|
Kaitaniemi S, Grön K, Elovaara H, Salmi M, Jalkanen S, Elima K. Functional modulation of vascular adhesion protein-1 by a novel splice variant. PLoS One 2013; 8:e54151. [PMID: 23349812 PMCID: PMC3548902 DOI: 10.1371/journal.pone.0054151] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 12/07/2012] [Indexed: 12/22/2022] Open
Abstract
Vascular Adhesion Protein-1 (VAP-1) is an endothelial adhesion molecule belonging to the primary amine oxidases. Upon inflammation it takes part in the leukocyte extravasation cascade facilitating transmigration of leukocytes into the inflamed tissue. Screening of a human lung cDNA library revealed the presence of an alternatively spliced shorter transcript of VAP-1, VAP-1Δ3. Here, we have studied the functional and structural characteristics of VAP-1Δ3, and show that the mRNA for this splice variant is expressed in most human tissues studied. In comparison to the parent molecule this carboxy-terminally truncated isoform lacks several of the amino acids important in the formation of the enzymatic groove of VAP-1. In addition, the conserved His684, which takes part in coordinating the active site copper, is missing from VAP-1Δ3. Assays using the prototypic amine substrates methylamine and benzylamine demonstrated that VAP-1Δ3 is indeed devoid of the semicarbazide-sensitive amine oxidase (SSAO) activity characteristic to VAP-1. When VAP-1Δ3-cDNA is transfected into cells stably expressing VAP-1, the surface expression of the full-length molecule is reduced. Furthermore, the SSAO activity of the co-transfectants is diminished in comparison to transfectants expressing only VAP-1. The observed down-regulation of both the expression and enzymatic activity of VAP-1 may result from a dominant-negative effect caused by heterodimerization between VAP-1 and VAP-1Δ3, which was detected in co-immunoprecipitation studies. This alternatively spliced transcript adds thus to the repertoire of potential regulatory mechanisms through which the cell-surface expression and enzymatic activity of VAP-1 can be modulated.
Collapse
Affiliation(s)
- Sam Kaitaniemi
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Microbiology and Immunology, University of Turku, Turku, Finland
| | - Kirsi Grön
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Heli Elovaara
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Microbiology and Immunology, University of Turku, Turku, Finland
| | - Kati Elima
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Drawnel FM, Wachten D, Molkentin JD, Maillet M, Aronsen JM, Swift F, Sjaastad I, Liu N, Catalucci D, Mikoshiba K, Hisatsune C, Okkenhaug H, Andrews SR, Bootman MD, Roderick HL. Mutual antagonism between IP(3)RII and miRNA-133a regulates calcium signals and cardiac hypertrophy. J Cell Biol 2012; 199:783-98. [PMID: 23166348 PMCID: PMC3514786 DOI: 10.1083/jcb.201111095] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 10/25/2012] [Indexed: 11/22/2022] Open
Abstract
Inositol 1,4,5'-triphosphate receptor II (IP(3)RII) calcium channel expression is increased in both hypertrophic failing human myocardium and experimentally induced models of the disease. The ectopic calcium released from these receptors induces pro-hypertrophic gene expression and may promote arrhythmias. Here, we show that IP(3)RII expression was constitutively restrained by the muscle-specific miRNA, miR-133a. During the hypertrophic response to pressure overload or neurohormonal stimuli, miR-133a down-regulation permitted IP(3)RII levels to increase, instigating pro-hypertrophic calcium signaling and concomitant pathological remodeling. Using a combination of in vivo and in vitro approaches, we demonstrated that IP(3)-induced calcium release (IICR) initiated the hypertrophy-associated decrease in miR-133a. In this manner, hypertrophic stimuli that engage IICR set a feed-forward mechanism in motion whereby IICR decreased miR-133a expression, further augmenting IP(3)RII levels and therefore pro-hypertrophic calcium release. Consequently, IICR can be considered as both an initiating event and a driving force for pathological remodeling.
Collapse
Affiliation(s)
- Faye M. Drawnel
- Babraham Institute, Babraham, Cambridge CB22 3AT, England, UK
| | - Dagmar Wachten
- Babraham Institute, Babraham, Cambridge CB22 3AT, England, UK
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, 53175 Bonn, Germany
| | - Jeffery D. Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Marjorie Maillet
- Department of Pediatrics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Faculty of Medicine, Oslo University Hospital, 0407 Oslo, Norway
- Bjørknes College, 0456 Oslo, Norway
| | - Fredrik Swift
- Institute for Experimental Medical Research, Faculty of Medicine, Oslo University Hospital, 0407 Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Faculty of Medicine, Oslo University Hospital, 0407 Oslo, Norway
| | - Ning Liu
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390
| | - Daniele Catalucci
- Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
- Institute of Genetic and Biomedical Research, Milan Section, National Research Council, 20138 Milan, Italy
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama 531-0198, Japan
| | - Chihiro Hisatsune
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama 531-0198, Japan
| | | | | | | | - H. Llewelyn Roderick
- Babraham Institute, Babraham, Cambridge CB22 3AT, England, UK
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| |
Collapse
|
18
|
Xu Z, Ji G, Shen J, Wang X, Zhou J, Li L. SOX9 and myocardin counteract each other in regulating vascular smooth muscle cell differentiation. Biochem Biophys Res Commun 2012; 422:285-90. [PMID: 22580282 DOI: 10.1016/j.bbrc.2012.04.149] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 04/26/2012] [Indexed: 02/06/2023]
Abstract
Transdifferentiation of vascular smooth muscle cells (VSMC) into chondrogenic cells contributes significantly to vascular calcification during the pathogenesis of atherosclerosis. However, the transcriptional mechanisms that control such phenotypic switch remain unclear. This process is characterized by the induction of Sox9 and Col2a1 genes accompanied by the repression of myocardin (Myocd) and SMC differentiation markers such as SM22, SM α-actin and SM-MHC. Here we explore the regulatory role of SOX9, the master regulator for chondrogenesis, in modulating SMC marker gene expression. qRT-PCR and luciferase assays show that over-expression of SOX9 inhibits SMC gene transcription and promoter activities induced by myocardin, the master regulator of smooth muscle differentiation. Such suppression is independent of the CArG box in the SMC promoters but dependent on myocardin. EMSA assay further shows that SOX9 neither participates in SRF (serum response factor) binding to the CArG box nor interacts with SRF, while co-immunoprecipitation demonstrates an association of SOX9 with myocardin. Conversely, myocardin suppresses SOX9-mediated chondrogenic gene Col2a1 expression. These findings provide the first mechanistic insights into the important regulatory role of SOX9 and myocardin in controlling the transcription program during SMC transdifferentiation into chondrocytes.
Collapse
Affiliation(s)
- Zhonghui Xu
- Department of Internal Medicine, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
19
|
Wong J, Zhang J, Yanagawa B, Luo Z, Yang X, Chang J, McManus B, Luo H. Cleavage of serum response factor mediated by enteroviral protease 2A contributes to impaired cardiac function. Cell Res 2012; 22:360-71. [PMID: 21769134 PMCID: PMC3271589 DOI: 10.1038/cr.2011.114] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 04/12/2011] [Accepted: 05/04/2011] [Indexed: 11/09/2022] Open
Abstract
Enteroviral infection can lead to dilated cardiomyopathy (DCM), which is a major cause of cardiovascular mortality worldwide. However, the pathogenetic mechanisms have not been fully elucidated. Serum response factor (SRF) is a cardiac-enriched transcription regulator controlling the expression of a variety of target genes, including those involved in the contractile apparatus and immediate early response, as well as microRNAs that silence the expression of cardiac regulatory factors. Knockout of SRF in the heart results in downregulation of cardiac contractile gene expression and development of DCM. The goal of this study is to understand the role of SRF in enterovirus-induced cardiac dysfunction and progression to DCM. Here we report that SRF is cleaved following enteroviral infection of mouse heart and cultured cardiomyocytes. This cleavage is accompanied by impaired cardiac function and downregulation of cardiac-specific contractile and regulatory genes. Further investigation by antibody epitope mapping and site-directed mutagenesis demonstrates that SRF cleavage occurs at the region of its transactivation domain through the action of virus-encoded protease 2A. Moreover, we demonstrate that cleavage of SRF dissociates its transactivation domain from DNA-binding domain, resulting in the disruption of SRF-mediated gene transactivation. In addition to loss of functional SRF, finally we report that the N-terminal fragment of SRF cleavage products can also act as a dominant-negative transcription factor, which likely competes with the native SRF for DNA binding. Our results suggest a mechanism by which virus infection impairs heart function and may offer a new therapeutic strategy to ameliorate myocardial damage and progression to DCM.
Collapse
Affiliation(s)
- Jerry Wong
- James Hogg iCAPTURE Centre, Providence Heart + Lung Institute, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y, Canada
| | - Jingchun Zhang
- James Hogg iCAPTURE Centre, Providence Heart + Lung Institute, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y, Canada
| | - Bobby Yanagawa
- James Hogg iCAPTURE Centre, Providence Heart + Lung Institute, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y, Canada
| | - Zongshu Luo
- James Hogg iCAPTURE Centre, Providence Heart + Lung Institute, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y, Canada
| | - Xiangsheng Yang
- Center for Molecular Development and Disease, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Jiang Chang
- Center for Molecular Development and Disease, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Bruce McManus
- James Hogg iCAPTURE Centre, Providence Heart + Lung Institute, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y, Canada
| | - Honglin Luo
- James Hogg iCAPTURE Centre, Providence Heart + Lung Institute, St Paul's Hospital and Department of Pathology and Laboratory Medicine, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y, Canada
| |
Collapse
|
20
|
Rensen S, Doevendans P, van Eys G. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J 2011; 15:100-8. [PMID: 17612668 PMCID: PMC1847757 DOI: 10.1007/bf03085963] [Citation(s) in RCA: 672] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vascular smooth muscle cells can perform both contractile and synthetic functions, which are associated with and characterised by changes in morphology, proliferation and migration rates, and the expression of different marker proteins. The resulting phenotypic diversity of smooth muscle cells appears to be a function of innate genetic programmes and environmental cues, which include biochemical factors, extracellular matrix components, and physical factors such as stretch and shear stress. Because of the diversity among smooth muscle cells, blood vessels attain the flexibility that is necessary to perform efficiently under different physiological and pathological conditions. In this review, we discuss recent literature demonstrating the extent and nature of smooth muscle cell diversity in the vascular wall and address the factors that affect smooth muscle cell phenotype. (Neth Heart J 2007;15:100-8.).
Collapse
Affiliation(s)
- S.S.M. Rensen
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands
| | - P.A.F.M. Doevendans
- Department of Cardiology, Heart Lung Centre Utrecht, Interuniversity Cardiology Institute, the Netherlands
| | - G.J.J.M. van Eys
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands
| |
Collapse
|
21
|
Sarkar A, Zhang M, Liu SH, Sarkar S, Brunicardi FC, Berger DH, Belaguli NS. Serum response factor expression is enriched in pancreatic β cells and regulates insulin gene expression. FASEB J 2011; 25:2592-603. [PMID: 21525490 DOI: 10.1096/fj.10-173757] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Serum response factor (SRF) is an essential regulator of myogenic and neurogenic genes and the ubiquitously expressed immediate-early genes. The purpose of this study is to determine SRF expression pattern in murine pancreas and examine the role of SRF in pancreatic gene expression. Immunohistochemical analysis of wild-type pancreas and LacZ staining of pancreas from SRF LacZ knock-in animals showed that SRF expression is restricted to β cells. SRF bound to the rat insulin promoter II (RIP II) serum response element, an element conserved in both rat I and murine I and II insulin promoters. SRF activated RIP II, and SRF binding to RIP II and the exon 5-encoded 64-aa subdomain of SRF was required for this activation. Transient or stable knockdown of SRF leads to down-regulation of insulin gene expression, suggesting that SRF is required for insulin gene expression. Further, SRF physically interacted with the pancreas and duodenum homeobox-1 (Pdx-1) and synergistically activated RIP II. Elevated glucose concentration down-regulated SRF binding to RIP II SRE, and this down-regulation was associated with decreased RIP II activity and increased SRF phosphorylation on serine 103. Together, our results demonstrate that SRF is a glucose concentration-sensitive regulator of insulin gene expression.
Collapse
Affiliation(s)
- Aloke Sarkar
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Perera S, Holt MR, Mankoo BS, Gautel M. Developmental regulation of MURF ubiquitin ligases and autophagy proteins nbr1, p62/SQSTM1 and LC3 during cardiac myofibril assembly and turnover. Dev Biol 2011; 351:46-61. [PMID: 21185285 PMCID: PMC3047806 DOI: 10.1016/j.ydbio.2010.12.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 02/07/2023]
Abstract
The striated muscle-specific tripartite motif (TRIM) proteins TRIM63/MURF1, TRIM55/MURF2 and TRIM54/MURF3 can function as ubiquitin E3 ligases in ubiquitin-mediated muscle protein turnover. Despite their well-characterised roles in muscle atrophy, the dynamics of MURF expression in the development and early postnatal adaptation of striated muscle is largely unknown. Here, we show that MURF2 is expressed at the very onset of mouse cardiac differentiation at embryonic day 8.5, and represents a sensitive marker for differentiating myocardium. During cardiac development, expression shifts from the 50 kDa to the 60 kDa A-isoform, which dominates postnatally. In contrast, MURF1 shows strong postnatal upregulation and MURF3 is not significantly expressed before birth. MURF2 expression parallels that of the autophagy-associated proteins LC3, p62/SQSTM1 and nbr1. SiRNA knockdown of MURF2 in neonatal rat cardiomyocytes disrupts posttranslational microtubule modification and myofibril assembly, and is only partly compensated by upregulation of MURF3 but not MURF1. Knockdown of both MURF2 and MURF3 severely disrupts the formation of ordered Z- and M-bands, likely by perturbed tubulin dynamics. These results suggest that ubiquitin-mediated protein turnover and MURF2 in particular play an unrecognised role in the earliest steps of heart muscle differentiation, and that partial complementation of MURF2 deficiency is afforded by MURF3.
Collapse
Affiliation(s)
| | | | | | - Mathias Gautel
- King's College London BHF Centre of Research Excellence, Randall Division for Cell and Molecular Biophysics and Cardiovascular Division, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
23
|
Sun Q, Taurin S, Sethakorn N, Long X, Imamura M, Wang DZ, Zimmer WE, Dulin NO, Miano JM. Myocardin-dependent activation of the CArG box-rich smooth muscle gamma-actin gene: preferential utilization of a single CArG element through functional association with the NKX3.1 homeodomain protein. J Biol Chem 2009; 284:32582-90. [PMID: 19797053 DOI: 10.1074/jbc.m109.033910] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serum response factor (SRF) is a ubiquitously expressed transcription factor that binds a 10-bp element known as the CArG box, located in the proximal regulatory region of hundreds of target genes. SRF activates target genes in a cell- and context-dependent manner by assembling unique combinations of cofactors over CArG elements. One particularly strong SRF cofactor, myocardin (MYOCD), acts as a component of a molecular switch for smooth muscle cell (SMC) differentiation by activating cytoskeletal and contractile genes harboring SRF-binding CArG elements. Here we report that the human ACTG2 promoter, containing four conserved CArG elements, displays SMC-specific basal activity and is highly induced in the presence of MYOCD. Stable transfection of a non-SMC cell type with Myocd elicits elevations in endogenous Actg2 mRNA. Gel shift and luciferase assays reveal a strong bias for MYOCD-dependent transactivation through CArG2 of the human ACTG2 promoter. Substitution of CArG2 with other CArGs, including a consensus CArG element, fails to reconstitute full MYOCD-dependent ACTG2 promoter stimulation. Mutation of an adjacent binding site for NKX3.1 reduces MYOCD-dependent transactivation of the ACTG2 promoter. Co-immunoprecipitation, glutathione S-transferase pulldown, and luciferase assays show a physical and functional association between MYOCD and NKX3.1; no such functional relationship is evident with the related NKX2.5 transcription factor despite its interaction with MYOCD. These results demonstrate the ability of MYOCD to discriminate among several juxtaposed CArG elements, presumably through its novel partnership with NKX3.1, to optimally transactivate the human ACTG2 promoter.
Collapse
Affiliation(s)
- Qiang Sun
- Aab Cardiovascular Research Institute of the University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Airway smooth muscle (SM) develops from local mesenchymal cells located around the tips of growing epithelial buds. These cells gradually displace from distal to proximal position alongside the bronchial tree, elongate, and begin to synthesize SM-specific proteins. Mechanical tension (either generated by cell spreading/elongation or stretch), as well as epithelial paracrine factors, regulates the process of bronchial myogenesis. The specific roles of many of these paracrine factors during normal lung development are currently unknown. It is also unknown how and if mechanical and paracrine signals integrate into a common myogenic pathway. Furthermore, as with vascular SM and other types of visceral SM, we are just beginning to elucidate the intracellular signaling pathways and the genetic program that controls lung myogenesis. Here we present what we have learned so far about the embryogenesis of bronchial muscle.
Collapse
|
25
|
Zhang A, David JJ, Subramanian SV, Liu X, Fuerst MD, Zhao X, Leier CV, Orosz CG, Kelm RJ, Strauch AR. Serum response factor neutralizes Pur alpha- and Pur beta-mediated repression of the fetal vascular smooth muscle alpha-actin gene in stressed adult cardiomyocytes. Am J Physiol Cell Physiol 2008; 294:C702-14. [PMID: 18344281 DOI: 10.1152/ajpcell.00173.2007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mouse hearts subjected to repeated transplant surgery and ischemia-reperfusion injury develop substantial interstitial and perivascular fibrosis that was spatially associated with dysfunctional activation of fetal smooth muscle alpha-actin (SM alpha A) gene expression in graft ventricular cardiomyocytes. Compared with cardiac fibroblasts in which nuclear levels of the Sp1 and Smad 2/3 transcriptional-activating proteins increased markedly after transplant injury, the most abundant SM alpha A gene-activating protein in cardiomyocyte nuclei was serum response factor (SRF). Additionally, cardiac intercalated discs in heart grafts contained substantial deposits of Pur alpha, an mRNA-binding protein and known negative modulator of SRF-activated SM alpha A gene transcription. Activation of fetal SM alpha A gene expression in perfusion-isolated adult cardiomyocytes was linked to elevated binding of a novel protein complex consisting of SRF and Pur alpha to a purine-rich DNA element in the SM alpha A promoter called SPUR, previously shown to be required for induction of SM alpha A gene transcription in injury-activated myofibroblasts. Increased SRF binding to SPUR DNA plus one of two nearby CArG box consensus elements was observed in SM alpha A-positive cardiomyocytes in parallel with enhanced Pur alpha:SPUR protein:protein interaction. The data suggest that de novo activation of the normally silent SM alpha A gene in reprogrammed adult cardiomyocytes is linked to elevated interaction of SRF with fetal-specific CArG and injury-activated SPUR elements in the SM alpha A promoter as well as the appearance of novel Pur alpha protein complexes in both the nuclear and cytosolic compartments of these cells.
Collapse
Affiliation(s)
- Aiwen Zhang
- Dept. of Physiology and Cell Biology, Davis Heart and Lung Research, Institute Biomedical Research Tower, Rm. 314, 460 West 12th Ave., The Ohio State Univ. College of Medicine, Columbus, OH 43210-1252, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Lightfoot DJ, Malone KM, Timmis JN, Orford SJ. Evidence for alternative splicing of MADS-box transcripts in developing cotton fibre cells. Mol Genet Genomics 2007; 279:75-85. [PMID: 17943315 DOI: 10.1007/s00438-007-0297-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 09/26/2007] [Indexed: 01/23/2023]
Abstract
The MADS-box family of genes encodes transcription factors that have widely ranging roles in diverse aspects of plant development. In this study, four cotton MADS-box cDNA clones of the type II (MIKC) class were isolated, with phylogenetic analysis indicating that the cotton sequences are of the AGAMOUS subclass. The corresponding transcripts were detected in developing cotton fibre cells as well as in whole ovule and flower tissue, with differential expression in stems, leaves and roots. Reverse transcription PCR showed extensive alternative splicing in one of the reactions, and 11 mRNAs of different intron/exon composition and length were characterised. Sequence differences between the transcripts indicated that they could not be derived from the same pre-mRNA and that the sequenced transcript pool was derived from two distinct MADS-box genes. Several of the alternatively spliced transcripts potentially encoded proteins with altered K-domains and/or C-terminal regions and the variant proteins may have altered cellular roles. This work is the first that describes MADS-box gene expression in elongating cotton fibres and adds to a growing body of evidence for the prevalence of alternative splicing in the expression of MADS-box and other genes.
Collapse
Affiliation(s)
- Damien J Lightfoot
- Discipline of Genetics, School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, SA, 5005, Australia
| | | | | | | |
Collapse
|
28
|
Zhang X, Azhar G, Huang C, Cui C, Zhong Y, Huck S, Wei JY. Alternative splicing and nonsense-mediated mRNA decay regulate gene expression of serum response factor. Gene 2007; 400:131-9. [PMID: 17629633 DOI: 10.1016/j.gene.2007.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 05/23/2007] [Accepted: 06/05/2007] [Indexed: 11/21/2022]
Abstract
Serum response factor (SRF) is an important transcription factor that regulates a variety of genes in many tissues during development, maturation and aging. The SRF protein also controls the expression of SRF target genes, including the SRF gene itself. However, it is incompletely established how SRF isoforms contribute to the regulation of SRF gene expression. In the present study, we report the identification of three novel SRF isoforms in human tissue. We found that one novel isoform, SRF-triangle up3, contained a premature termination codon (PTC), which was a target of nonsense-mediated mRNA decay (NMD). By contrast, the SRF-triangle up345 isoform protein was able to specifically bind to the serum response element, and to repress the SRF gene promoter activity. Therefore, we propose that SRF isoforms regulate expression of the SRF gene via two different mechanisms. One mechanism is to reduce the abundance of SRF transcripts via coupled alternative splicing and NMD, the other one is to regulate the SRF gene expression via a feedback mechanism in which the SRF isoform proteins bind to the SRF gene promoter region. Analysis of hundreds of SRF cDNA clones derived from human hearts of fetuses, young adults, old and very old individuals revealed that SRF isoform transcripts were increased in the human heart with advancing age. Our data indicate that the SRF isoforms were differentially expressed in the human versus mouse cardiac muscle. Alternative splicing and NMD likely maintain a delicate balance of SRF transcripts and/or proteins among the full-length SRF form and various SRF isoforms that are critical to the regulation of many SRF target genes, including the SRF gene itself.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Donald W. Reynolds Department of Geriatrics, University of Arkansas for Medical Sciences and Geriatric Research, Education, and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, United States
| | | | | | | | | | | | | |
Collapse
|
29
|
Asanoma K, Kato H, Yamaguchi S, Shin CH, Liu ZP, Kato K, Inoue T, Miyanari Y, Yoshikawa K, Sonoda K, Fukushima K, Wake N. HOP/NECC1, A Novel Regulator of Mouse Trophoblast Differentiation. J Biol Chem 2007; 282:24065-74. [PMID: 17576768 DOI: 10.1074/jbc.m701380200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Homeodomain-only protein/not expressed in choriocarcinoma clone 1 (HOP/NECC1) is a newly identified gene that modifies the expression of cardiac-specific genes and thereby regulates heart development. More recently, HOP/NECC1 was reported to be a suppressor of choriocarcinogenesis. Here, we examined the temporal expression profile of HOP/NECC1 in wild-type mouse placenta. We found that E8.5-E9.5 wild-type placenta expressed HOP/NECC1 in the giant cell and spongiotrophoblast layers. HOP/NECC1 (-/-) placenta exhibited marked propagation of giant cell layers and, in turn reduction of spongiotrophoblast formation. We demonstrated SRF transcriptional activity increased in the differentiating trophoblasts and forced expression of SRF in a trophoblast stem (TS) cell line induces the differentiation into giant cells. Negative regulation of SRF (serum response factor) by the binding of HOP/NECC1 protein contributed at least in part to the generation of these placental defects. Gradual induction of HOP/NECC1 in response to differentiation stimuli may result in the decision to differentiate into a particular type of trophoblastic cell lineage and result in non-lethal defects shown by the HOP/NECC1 (-/-) placentas.
Collapse
Affiliation(s)
- Kazuo Asanoma
- Division of Molecular and Cell Therapeutics, Kyushu University, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chow N, Bell RD, Deane R, Streb JW, Chen J, Brooks A, Van Nostrand W, Miano JM, Zlokovic BV. Serum response factor and myocardin mediate arterial hypercontractility and cerebral blood flow dysregulation in Alzheimer's phenotype. Proc Natl Acad Sci U S A 2007; 104:823-8. [PMID: 17215356 PMCID: PMC1783398 DOI: 10.1073/pnas.0608251104] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cerebral angiopathy contributes to cognitive decline and dementia in Alzheimer's disease (AD) through cerebral blood flow (CBF) reductions and dysregulation. We report vascular smooth muscle cells (VSMC) in small pial and intracerebral arteries, which are critical for CBF regulation, express in AD high levels of serum response factor (SRF) and myocardin (MYOCD), two interacting transcription factors that orchestrate a VSMC-differentiated phenotype. Consistent with this finding, AD VSMC overexpressed several SRF-MYOCD-regulated contractile proteins and exhibited a hypercontractile phenotype. MYOCD overexpression in control human cerebral VSMC induced an AD-like hypercontractile phenotype and diminished both endothelial-dependent and -independent relaxation in the mouse aorta ex vivo. In contrast, silencing SRF normalized contractile protein content and reversed a hypercontractile phenotype in AD VSMC. MYOCD in vivo gene transfer to mouse pial arteries increased contractile protein content and diminished CBF responses produced by brain activation in wild-type mice and in two AD models, the Dutch/Iowa/Swedish triple mutant human amyloid beta-peptide (Abeta)-precursor protein (APP)- expressing mice and APPsw(+/-) mice. Silencing Srf had the opposite effect. Expression of SRF did not change in VSMC subjected to Alzheimer's neurotoxin, Abeta. Thus, SRF-MYOCD overexpression in small cerebral arteries appears to initiate independently of Abeta a pathogenic pathway mediating arterial hypercontractility and CBF dysregulation, which are associated with Alzheimer's dementia.
Collapse
Affiliation(s)
| | | | - Rashid Deane
- Frank P. Smith Laboratories for Neuroscience and Neurosurgical Research, Department of Neurosurgery
| | | | | | - Andrew Brooks
- Functional Genomics Core, Arthur Kornberg Medical Research Building, University of Rochester School of Medicine, 601 Elmwood Avenue, Rochester, NY 14642; and
| | | | | | - Berislav V. Zlokovic
- Frank P. Smith Laboratories for Neuroscience and Neurosurgical Research, Department of Neurosurgery
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
31
|
Zhang SX, Garcia-Gras E, Wycuff DR, Marriot SJ, Kadeer N, Yu W, Olson EN, Garry DJ, Parmacek MS, Schwartz RJ. Identification of Direct Serum-response Factor Gene Targets during Me2SO-induced P19 Cardiac Cell Differentiation. J Biol Chem 2005; 280:19115-26. [PMID: 15699019 DOI: 10.1074/jbc.m413793200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Serum-response factor (SRF) is an obligatory transcription factor, required for the formation of vertebrate mesoderm leading to the origin of the cardiovascular system. Protein A-TEV-tagged chromatin immunoprecipitation technology was used to collect direct SRF-bound gene targets from pluripotent P19 cells, induced by Me2SO treatment into an enriched cardiac cell population. From 242 sequenced DNA fragments, we identified 188 genomic DNA fragments as potential direct SRF targets that contain CArG boxes and CArG-like boxes. Of the 92 contiguous genes that were identified, a subgroup of 43 SRF targets was then further validated by co-transfection assays with SRF. Expression patterns of representative candidate genes were compared with the LacZ reporter expression activity of the endogenous SRF gene. According to the Unigene data base, 84% of the SRF target candidates were expressed, at least, in the heart. In SRF null embryonic stem cells, 81% of these SRF target candidates were greatly affected by the absence of SRF. Among these SRF-regulated genes, Raf1, Map4k4, and Bicc1 have essential roles in mesoderm formation. The 12 regulated SRF target genes, Mapk10 (JNK3), Txnl2, Azi2, Tera, Sema3a, Lrp4, Actc1, Myl3, Hspg2, Pgm2, Hif3a, and Asb5, have been implicated in cardiovascular formation, and the Ski and Hes6 genes have roles in muscle differentiation. SRF target genes related to cell mitosis and cycle, E2f5, Npm1, Cenpb, Rbbp6, and Scyl1, expressed in the heart tissue were differentially regulated in SRF null ES cells.
Collapse
Affiliation(s)
- Shu Xing Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Although the primary role of vascular smooth muscle cells (SMCs) is contraction, they exhibit extensive phenotypic diversity and plasticity during normal development, during repair of vascular injury, and in disease states. Results of recent studies indicate that there are unique as well as common transcriptional regulatory mechanisms that control expression of various SMC marker genes in distinct SMC subtypes, and that these mechanisms are complex and dynamic even at the single cell level. This article will review recent progress in our understanding of the transcriptional regulatory mechanisms involved in controlling expression of SMC marker genes with a particular focus on examination of processes that contribute to the phenotypic diversity of SMCs. In addition, because of considerable controversy in the literature regarding the relationship between phenotypically modulated SMCs and myofibroblasts, we will briefly consider both similarities and differences in regulation of gene expression between these cell types.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville 22908, USA
| | | |
Collapse
|
33
|
Kimura E, Hidaka K, Kida Y, Morisaki H, Shirai M, Araki K, Suzuki M, Yamamura KI, Morisaki T. Serine-arginine-rich nuclear protein Luc7l regulates myogenesis in mice. Gene 2005; 341:41-7. [PMID: 15474286 DOI: 10.1016/j.gene.2004.07.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2004] [Revised: 06/25/2004] [Accepted: 07/19/2004] [Indexed: 11/23/2022]
Abstract
Using a gene trap technique, we identified a murine homologue of the yeast LUC7-like gene (Luc7l), which is a serine-arginine-rich protein (SR protein) that localizes in the nucleus through its arginine-serine-rich domain (RS domain) at the C-terminus and shows a speckled distribution pattern. Although its transcripts are widely expressed in embryos and adults, they are rarely detected in adult skeletal muscle, and Luc7l expression was found to be negatively regulated during the course of development of limb skeletal muscle, as well as during in vitro differentiation of the myoblast cell lines Sol8 and C2C12. We also demonstrated that forced expression of Luc7l protein inhibited myogenesis in vitro. Based on our results, Luc7l is thought to play an important role in the regulation of muscle differentiation.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Western
- COS Cells
- Cell Differentiation/genetics
- Cell Differentiation/physiology
- Cell Line
- Chlorocebus aethiops
- Down-Regulation/genetics
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Male
- Mice
- Mice, Mutant Strains
- Microscopy, Fluorescence
- Muscle Development/genetics
- Muscle Development/physiology
- Muscle, Skeletal/embryology
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/metabolism
- Myoblasts/cytology
- Myoblasts/metabolism
- Nuclear Proteins/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Transfection
Collapse
Affiliation(s)
- E Kimura
- Department of Bioscience, National Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vlahopoulos S, Zimmer WE, Jenster G, Belaguli NS, Balk SP, Brinkmann AO, Lanz RB, Zoumpourlis VC, Schwartz RJ. Recruitment of the androgen receptor via serum response factor facilitates expression of a myogenic gene. J Biol Chem 2004; 280:7786-92. [PMID: 15623502 DOI: 10.1074/jbc.m413992200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Androgen receptor (AR) induced precocious myogenesis in culture and myogenic specified gene activity. Increased levels of AR expression in replicating C2C12 myoblasts stimulated fusion into post-differentiated multinucleated myotubes and the appearance of skeletal alpha-actin transcripts, even in the absence of ligand. Furthermore, AR activated the skeletal alpha-actin promoter, which lacks GRE sites, in co-transfected C2C12 cells. AR co-activation of the skeletal alpha-actin promoter required co-expressed full-length serum response factor (SRF). In vitro, AR associated with SRF and was recruited by SRF to a alpha-actin promoter SRF binding site. Our data suggest that AR is capable of activating myogenic genes devoid of consensus AR binding sites via its recruitment by the myogenic enriched transcription factor, SRF.
Collapse
Affiliation(s)
- Spiros Vlahopoulos
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Patten LC, Belaguli NS, Baek MJ, Fagan SP, Awad SS, Berger DH. Serum response factor is alternatively spliced in human colon cancer. J Surg Res 2004; 121:92-100. [PMID: 15313381 DOI: 10.1016/j.jss.2004.02.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2003] [Indexed: 10/26/2022]
Abstract
BACKGROUND Serum response factor (SRF) is a transcription factor that plays an important role in cellular differentiation and cell cycle regulation. SRF function is regulated in part by alternative splicing. Little is known about the expression or role of these alternatively spliced isoforms during tumorigenesis. We hypothesized that there is a change in the splice variants during intestinal tumorigenesis and that this change promotes the tumor phenotype. MATERIALS AND METHODS SRF expression was determined by Western blotting of benign intestinal cells and human colon cancer cell lines. To determine the effect of alternative splicing of SRF on intestinal growth and proliferation, the predominant alternatively spliced isoform of SRF that we identified in colon cancer cells, SRFDelta5, was transfected into IEC-6 cells. IEC-6 and IEC-6SRFDelta5 cells were plated and cell numbers were determined at four time points. RESULTS Western blotting demonstrates that full-length SRF is the predominant form of SRF in rat IEC-6 cells, normal human colonic mucosa, and HT-29 cells, derived from a well-differentiated human colonic adenocarcinoma. In the colon cancer cell lines derived from poorly differentiated tumors (WiDr, HCT 116, LoVo, and SW480), SRFDelta5 is the predominant isoform expressed. There was a significant increase in cell survival in IEC-6 cells transfected with SRFDelta5 compared to parental cells. CONCLUSION Our data demonstrate that an alternatively spliced isoform of SRF, SRFDelta5, is expressed in human colon cancer cell lines. Additionally, these data demonstrate that expression of SRFDelta5 may contribute to the tumor phenotype by affecting cell survival. This is the first study to document a change in expression of the alternatively spliced isoform of SRF in human malignancy.
Collapse
Affiliation(s)
- Lane C Patten
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, and The Houston Veterans Affairs Hospital, 2002 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 2004; 84:767-801. [PMID: 15269336 DOI: 10.1152/physrev.00041.2003] [Citation(s) in RCA: 2629] [Impact Index Per Article: 125.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The focus of this review is to provide an overview of the current state of knowledge of molecular mechanisms/processes that control differentiation of vascular smooth muscle cells (SMC) during normal development and maturation of the vasculature, as well as how these mechanisms/processes are altered in vascular injury or disease. A major challenge in understanding differentiation of the vascular SMC is that this cell can exhibit a wide range of different phenotypes at different stages of development, and even in adult organisms the cell is not terminally differentiated. Indeed, the SMC is capable of major changes in its phenotype in response to changes in local environmental cues including growth factors/inhibitors, mechanical influences, cell-cell and cell-matrix interactions, and various inflammatory mediators. There has been much progress in recent years to identify mechanisms that control expression of the repertoire of genes that are specific or selective for the vascular SMC and required for its differentiated function. One of the most exciting recent discoveries was the identification of the serum response factor (SRF) coactivator gene myocardin that appears to be required for expression of many SMC differentiation marker genes, and for initial differentiation of SMC during development. However, it is critical to recognize that overall control of SMC differentiation/maturation, and regulation of its responses to changing environmental cues, is extremely complex and involves the cooperative interaction of many factors and signaling pathways that are just beginning to be understood. There is also relatively recent evidence that circulating stem cell populations can give rise to smooth muscle-like cells in association with vascular injury and atherosclerotic lesion development, although the exact role and properties of these cells remain to be clearly elucidated. The goal of this review is to summarize the current state of our knowledge in this area and to attempt to identify some of the key unresolved challenges and questions that require further study.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Arteriosclerosis/genetics
- Cell Differentiation
- Cellular Senescence
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Vascular Diseases/genetics
- Vascular Diseases/metabolism
- Vascular Diseases/pathology
Collapse
Affiliation(s)
- Gary K Owens
- Dept. of Molecular Physiology and Biological Physics, Univ. of Virginia School of Medicine, 415 Lane Rd., Medical Research Building 5, Rm. 1220, PO Box 801394, Charlottesville, VA 22908, USA.
| | | | | |
Collapse
|
38
|
Kawai-Kowase K, Sato H, Oyama Y, Kanai H, Sato M, Doi H, Kurabayashi M. Basic fibroblast growth factor antagonizes transforming growth factor-beta1-induced smooth muscle gene expression through extracellular signal-regulated kinase 1/2 signaling pathway activation. Arterioscler Thromb Vasc Biol 2004; 24:1384-90. [PMID: 15217807 DOI: 10.1161/01.atv.0000136548.17816.07] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Transforming growth factor-beta1 (TGFbeta1) and fibroblast growth factor (FGF) families play a pivotal role during vascular development and in the pathogenesis of vascular disease. However, the interaction of intracellular signaling evoked by each of these growth factors is not well understood. The present study was undertaken to examine the molecular mechanisms that mediate the effects of TGFbeta1 and basic FGF (bFGF) on smooth muscle cell (SMC) gene expression. METHODS AND RESULTS TGFbeta1 induction of SMC gene expression, including smooth muscle protein 22-alpha (SM22alpha) and smooth muscle alpha-actin, was examined in the pluripotent 10T1/2 cells. Marked increase in these mRNA levels by TGFbeta1 was inhibited by c-Src-tyrosine kinase inhibitors and protein synthesis inhibitor cycloheximide. Functional studies with deletion and site-directed mutation analysis of the SM22alpha promoter demonstrated that TGFbeta1 activated the SM22alpha promoter through a CC(A/T-rich)6GG (CArG) box, which serves as a serum response factor (SRF)-binding site. TGFbeta1 increased SRF expression through an increase in transcription of the SRF gene. In the presence of bFGF, TGFbeta1 induction of SMC marker gene expression was significantly attenuated. Transient transfection assays showed that bFGF significantly suppressed induction of the SM22alpha promoter-driven luciferase activity by TGFbeta1, whereas bFGF had no effects on the TGFbeta1-mediated increase in SRF expression and SRF:DNA binding activity. Mitogen-activated protein kinase kinase-1 (MEK1) inhibitor PD98059 abrogated the bFGF-mediated suppression of TGFbeta1-induced SMC gene expression. CONCLUSIONS Our data suggest that bFGF-induced MEK/extracellular signal-regulated kinase signaling plays an antagonistic role in TGFbeta1-induced SMC gene expression through suppression of the SRF function. These data indicate that opposing effects of bFGF and TGFbeta1 on SMC gene expression control the phenotypic plasticity of SMCs.
Collapse
MESH Headings
- Actins/biosynthesis
- Actins/genetics
- Animals
- Aorta
- Calcium-Binding Proteins/biosynthesis
- Calcium-Binding Proteins/genetics
- Cell Differentiation/drug effects
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Cycloheximide/pharmacology
- DNA/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression Regulation/drug effects
- MAP Kinase Kinase 1/antagonists & inhibitors
- MAP Kinase Kinase 1/physiology
- Mice
- Mice, Inbred C3H
- Microfilament Proteins/biosynthesis
- Microfilament Proteins/genetics
- Mitogen-Activated Protein Kinase 1/physiology
- Mitogen-Activated Protein Kinase 3/physiology
- Muscle Proteins/biosynthesis
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/cytology
- Mutagenesis, Site-Directed
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Promoter Regions, Genetic/drug effects
- Protein Binding
- Protein Kinase Inhibitors/pharmacology
- Protein Synthesis Inhibitors/pharmacology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Recombinant Proteins/pharmacology
- Serum Response Element/drug effects
- Serum Response Factor/metabolism
- Serum Response Factor/pharmacology
- Signal Transduction/drug effects
- Transcription, Genetic/drug effects
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta1
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/physiology
- Calponins
Collapse
Affiliation(s)
- Keiko Kawai-Kowase
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-Maebashi, Gunma, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Wycuff DR, Yanites HL, Marriott SJ. Identification of a functional serum response element in the HTLV-I LTR. Virology 2004; 324:540-53. [PMID: 15207639 DOI: 10.1016/j.virol.2004.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2004] [Revised: 03/15/2004] [Accepted: 04/07/2004] [Indexed: 11/19/2022]
Abstract
In response to various mitogenic signals, serum response factor (SRF) activates cellular gene expression after binding to its cognate target sequence (CArG box) located within a serum response element (SRE). SRF is particularly important in T cell activation, and we now report that SRF activates basal transcription from the human T-cell leukemia virus-I (HTLV-I) long terminal repeat (LTR). A DNA element, with similarity to the consensus cellular CArG box found in the c-fos promoter centered approximately 120 base pairs upstream from the viral transcription start site, has been identified and named the vCArG box. SRF activation of gene expression from the LTR was localized to the vCArG box, and mutation of this site abolished SRF responsiveness. An oligonucleotide probe containing the vCArG box bound purified SRF, and a complex formed on this probe with nuclear extract was supershifted by anti-SRF antibody. Moreover, a biotinylated probe containing the vCArG box bound SRF in avidin-biotin pull-down assays. Quantitative binding analysis yielded nanomolar affinities for both the viral and cellular CArG boxes. Chromatin immunoprecipitation experiments demonstrated that SRF is resident on the HTLV-I LTR in vivo. These data identify a functional serum response element in the HTLV-I LTR and suggest that SRF may play an important role in regulating basal HTLV-I gene expression in early infection and reactivation from latency.
Collapse
Affiliation(s)
- Diane R Wycuff
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | |
Collapse
|
40
|
Ferrario S, Busscher J, Franken J, Gerats T, Vandenbussche M, Angenent GC, Immink RGH. Ectopic expression of the petunia MADS box gene UNSHAVEN accelerates flowering and confers leaf-like characteristics to floral organs in a dominant-negative manner. THE PLANT CELL 2004; 16:1490-505. [PMID: 15155884 PMCID: PMC490041 DOI: 10.1105/tpc.019679] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Accepted: 03/25/2004] [Indexed: 05/18/2023]
Abstract
Several genes belonging to the MADS box transcription factor family have been shown to be involved in the transition from vegetative to reproductive growth. The Petunia hybrida MADS box gene UNSHAVEN (UNS) shares sequence similarity with the Arabidopsis thaliana flowering gene SUPPRESSOR OF OVEREXPRESSION OF CONSTANS1, is expressed in vegetative tissues, and is downregulated upon floral initiation and the formation of floral meristems. To understand the role of UNS in the flowering process, knockout mutants were identified and UNS was expressed ectopically in petunia and Arabidopsis. No phenotype was observed in petunia plants in which UNS was disrupted by transposon insertion, indicating that its function is redundant. Constitutive expression of UNS leads to an acceleration of flowering and to the unshaven floral phenotype, which is characterized by ectopic trichome formation on floral organs and conversion of petals into organs with leaf-like features. The same floral phenotype, accompanied by a delay in flowering, was obtained when a truncated version of UNS, lacking the MADS box domain, was introduced. We demonstrated that the truncated protein is not translocated to the nucleus. Using the overexpression approach with both the full-length and the nonfunctional truncated UNS protein, we could distinguish between phenotypic alterations because of a dominant-negative action of the protein and because of its native function in promoting floral transition.
Collapse
Affiliation(s)
- Silvia Ferrario
- Business Unit Bioscience, Plant Research International, 6700 AA, Wageningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Mechanical force is a critical modulator of smooth muscle (SM) function and gene expression. Very little is known, however, about its contribution to SM myogenesis. This review presents and discusses what has been learned about the role of mechanical force in inducing SM myogenesis and some of the signaling mechanisms involved in this process.
Collapse
Affiliation(s)
- Sandhya Jakkaraju
- Department of Pathology, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
42
|
Yang Y, Zhe X, Phan SH, Ullenbruch M, Schuger L. Involvement of serum response factor isoforms in myofibroblast differentiation during bleomycin-induced lung injury. Am J Respir Cell Mol Biol 2003; 29:583-90. [PMID: 12777247 DOI: 10.1165/rcmb.2002-0315oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Serum response factor (SRF) is a transcription factor essential for smooth muscle (SM) myogenesis. Its role in myofibroblast differentiation is, however, unknown. We studied the expression and the localization of SRF in bleomycin-induced pulmonary fibrosis, where myofibroblasts are abundant. We found that SRF levels were upregulated in bleomycin-exposed mouse lungs mainly due to de novo synthesis of SRFDelta5, a less myogenic SRF isoform. Before myofibroblast differentiation, SRF/SRFDelta5 was immunolocalized mostly in the cytoplasm of scattered fibroblasts at lesion sites. With the development of myofibroblasts, however, SRF/SRFDelta5 was found in myofibroblast nuclei. cDNA array analysis showed that SRFDelta5 and SRF induced expression of transforming growth factor-beta1, a critical factor in myofibroblast differentiation. This was accompanied by de novo expression of several inflammatory cell-specific mRNAs. The latter was confirmed by reverse transcriptase-polymerase chain reaction. Treatment of lung fibroblasts with tumor necrosis factor-alpha, which is produced early in the bleomycin model, induced SRFDelta5 expression and SRF/SRFDelta5 cytoplasmic accumulation, whereas addition of transforming growth factor-beta1 caused SRF/SRFDelta5 nuclear translocation followed by SM alpha-actin synthesis. Interleukin-4, another cytokine involved in myofibroblast differentiation, did not affect SRF or induce SRFDelta5 expression. Our studies therefore suggested a new mechanism whereby SRF and SRFDelta5 contribute to the emergence of myofibroblasts in lung injury and fibrosis.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pathology, Wayne State University School of Medicine, 540 E. Canfield St., Rm. 9248, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
43
|
Chang YF, Wei J, Liu X, Chen YH, Layne MD, Yet SF. Identification of a CArG-independent region of the cysteine-rich protein 2 promoter that directs expression in the developing vasculature. Am J Physiol Heart Circ Physiol 2003; 285:H1675-83. [PMID: 12791591 DOI: 10.1152/ajpheart.00165.2003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cysteine-rich protein (CRP)2 is a member of the LIM-only CRP family that is expressed in vascular smooth muscle cells (VSMC). To gain insight into the transcription of CSRP2 (gene name for CRP2) in VSMC, we analyzed the 5'-flanking sequence of the CSRP2 gene. We showed previously that 4,855 bp of the 5'-flanking sequence of the CSRP2 gene directed lacZ reporter gene expression, primarily in the VSMC of transgenic mice. To further define the regulatory sequences important for CSRP2 expression in VSMC, a series of promoter constructs containing deletions of the 5'-flanking sequence upstream of a nuclear-localized lacZ reporter gene were generated and analyzed. Similar to that observed in the -4855CSRP2-lacZ mice, beta-galactosidase reporter activity was detected in the developing great vessels, aorta, intersegmental arteries, umbilical vessels, endocardial cushions, and neural tube in the -3513-, -2663-, -795-, and -664CSRP2-lacZ lines. However, an internal deletion of bp -573 to -550 abolished the vascular, but not the neural tube, staining. Interestingly, no CArG box [CC(A/T)6GG] was present in the -795-bp fragment. Cotransfection experiments showed that dominant-negative serum response factor (SRF) did not repress CSRP2 promoter activity, which was different from the repressive effect of dominant-negative SRF on the SM22 alpha promoter. Our data suggest the presence of a VSMC-specific element(s) within bp -573 to -550 of the CSRP2 5'-flanking sequence; however, in contrast to many other smooth muscle genes, transcriptional regulation of the CSRP2 gene is not dependent on SRF.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Base Sequence/genetics
- Blood Vessels/embryology
- Blood Vessels/growth & development
- Blood Vessels/metabolism
- Cell Cycle Proteins
- Cells, Cultured
- DNA-Binding Proteins/physiology
- Embryo, Mammalian/metabolism
- Embryonic and Fetal Development
- Gene Expression
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B
- Male
- Mice
- Mice, Transgenic
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/physiology
- Protein Structure, Tertiary/genetics
- Proteins
- Rats
- Rats, Sprague-Dawley
- Repressor Proteins/physiology
- Ribonucleoproteins
- Transcription Factors
- Transgenes
Collapse
Affiliation(s)
- Yung-Fu Chang
- Pulmonary and Critical Care Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
44
|
Harding EW, Tang W, Nichols KW, Fernandez DE, Perry SE. Expression and maintenance of embryogenic potential is enhanced through constitutive expression of AGAMOUS-Like 15. PLANT PHYSIOLOGY 2003; 133:653-63. [PMID: 14512519 PMCID: PMC219041 DOI: 10.1104/pp.103.023499] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2003] [Revised: 05/06/2003] [Accepted: 07/01/2003] [Indexed: 05/18/2023]
Abstract
The MADS domain protein AGL15 (AGAMOUS-Like 15) has been found to preferentially accumulate in angiosperm tissues derived from double fertilization (i.e. the embryo, suspensor, and endosperm) and in apomictic, somatic, and microspore embryos. Localization to the nuclei supports a role in gene regulation during this phase of the life cycle. To test whether AGL15 is involved in the promotion and maintenance of embryo identity, the embryogenic potential of transgenic plants that constitutively express AGL15 was assessed. Expression of AGL15 was found to enhance production of secondary embryos from cultured zygotic embryos, and constitutive expression led to long-term maintenance of development in this mode. Ectopic accumulation of AGL15 also promoted somatic embryo formation after germination from the shoot apical meristem of seedlings in culture. These results indicate that AGL15 is involved in support of development in an embryonic mode.
Collapse
Affiliation(s)
- Ellen W Harding
- Department of Agronomy, University of Kentucky, 1405 Veterans Drive, Lexington, KY 40546-0312, USA
| | | | | | | | | |
Collapse
|
45
|
Camoretti-Mercado B, Dulin NO, Solway J. Serum response factor function and dysfunction in smooth muscle. Respir Physiol Neurobiol 2003; 137:223-35. [PMID: 14516728 DOI: 10.1016/s1569-9048(03)00149-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tight control of smooth muscle cell (SM) proliferation, differentiation, and apoptosis requires a balance between signaling and transcriptional events. Recent developments in vascular research revealed that serum response factor (SRF) function is important for the regulation of each of these processes. The cloning and characterization of several SM specific genes and the discovery that SRF is central for their expression fueled studies aimed at understanding the role of molecular partners including co-activators and co-repressors. Perturbations of pathways involving SRF are associated with abnormalities in the myogenic program and aberrant phenotypic consequences. Surprisingly, studies on airway SM have remained an underrepresented area of investigation. Our laboratory described a novel regulatory mechanism of SRF function in airway myocytes by modulation of its subcellular localization. This review summarizes current knowledge on the structure and function of this essential transcription factor as well different modes of regulating SRF expression and activity that are becoming key players in directing SM function in health and disease.
Collapse
Affiliation(s)
- Blanca Camoretti-Mercado
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S Maryland Avenue, MC6026, Chicago, IL 60637,USA.
| | | | | |
Collapse
|
46
|
Chang J, Wei L, Otani T, Youker KA, Entman ML, Schwartz RJ. Inhibitory cardiac transcription factor, SRF-N, is generated by caspase 3 cleavage in human heart failure and attenuated by ventricular unloading. Circulation 2003; 108:407-13. [PMID: 12874181 DOI: 10.1161/01.cir.0000084502.02147.83] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Knowledge about molecular mechanisms leading to heart failure is still limited, but reduced gene activities and modest activation of caspase 3 are hallmarks of end-stage heart failure. We postulated that serum response factor (SRF), a central cardiac transcription factor, might be a cleavage target for modest activated caspase 3, and this cleavage of SRF may play a dominant inhibitory role in propelling hearts toward failure. METHODS AND RESULTS We examined SRF protein levels from cardiac samples taken at the time of transplantation in 13 patients with end-stage heart failure and 7 normal hearts. Full-length SRF was markedly reduced and processed into 55- and 32-kDa subfragments in all failing hearts. SRF was intact in normal samples. In contrast, the hearts of 10 patients with left ventricular assist devices showed minimal SRF fragmentation. Specific antibodies to N- and C-terminal SRF sequences and site-directed mutagenesis revealed 2 alternative caspase 3 cleavage sites, so that 2 fragments were detected of each containing either the N- or C-terminal SRF. Expression of SRF-N, the 32-kDa fragment, in myogenic cells inhibited the transcriptional activity of alpha-actin gene promoters by 50% to 60%, which suggests that truncated SRF functioned as a dominant-negative transcription factor. CONCLUSIONS Caspase 3 activation in heart failure sequentially cleaved SRF and generated a dominant-negative transcription factor, which may explain the depression of cardiac-specific genes. Moreover, caspase 3 activation may be reversible in the failing heart with ventricular unloading.
Collapse
Affiliation(s)
- Jiang Chang
- Department of Molecular and Cellular Biology, Center for Cardiovascular Development, Methodist Hospital, Baylor College of Medicine, One Baylor Plaza, Houston, Tex 77030, USA
| | | | | | | | | | | |
Collapse
|
47
|
Carson JA, Culberson DE, Thompson RW, Fillmore RA, Zimmer W. Smooth muscle gamma-actin promoter regulation by RhoA and serum response factor signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1628:133-9. [PMID: 12890560 DOI: 10.1016/s0167-4781(03)00122-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Smooth muscle gamma-actin (SMGA) is both an early marker of smooth muscle cell differentiation, which demonstrates an expression pattern restricted to smooth muscle and the post meiotic spermatocyte. Serum response factor (SRF) DNA-binding is an important regulator of muscle differentiation, including SMGA expression during smooth muscle cell differentiation. RhoA, a low molecular weight GTPase protein, can regulate cardiac, skeletal, and smooth muscle differentiation through SRF-dependent mechanisms. This study's purpose was to examine RhoA expression during smooth muscle cell development, and determine if the SMGA promoter activity is sensitive to RhoA-mediated signaling through SRF. Additionally, the study identified the promoter regulation modifying SMGA expression by RhoA signaling. Western blot analysis of embryonic chick gizzard whole protein extracts during 5 to 14 days of development demonstrated a large induction of RhoA (10-fold) and beta1 integrin expression at day 8, which corresponds to the time SMGA expression and differentiation are occurring. Transient transfections in CV-1 fibroblast cells demonstrated that co-overexpression of SRF and RhoA could induce a 40-fold induction of -176 bp SMGA promoter activity. Mutational analysis demonstrated that serum response element (SRE)-1, but not SRE2, was necessary for RhoA/SRF activation of the SMGA promoter. Deletion analysis revealed that although SRE1 was necessary for SMGA promoter activation by RhoA and SRF, it was not sufficient, implicating a possible obligatory role of additional promoter sequences in the response. Overexpression of a mutated SRF protein that was unable to bind DNA demonstrated that the 40-fold RhoA/SRF activation was largely dependent on SRF binding to the SMGA promoter. Thus, as the SMGA promoter appears to be a target of RhoA-mediated transcriptional regulation, the uncovering of these signaling mechanisms effecting SMGA promoter activity should provide a regulatory paradigm that can then be examined during the regulation of other smooth muscle genes.
Collapse
Affiliation(s)
- James A Carson
- Integrative Muscle Biology Laboratory, Exercise Science Department, University of South Carolina, 1300 Wheat St., Columbia, SC 29208, USA.
| | | | | | | | | |
Collapse
|
48
|
Davis FJ, Gupta M, Camoretti-Mercado B, Schwartz RJ, Gupta MP. Calcium/calmodulin-dependent protein kinase activates serum response factor transcription activity by its dissociation from histone deacetylase, HDAC4. Implications in cardiac muscle gene regulation during hypertrophy. J Biol Chem 2003; 278:20047-58. [PMID: 12663674 DOI: 10.1074/jbc.m209998200] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serum response factor (SRF) plays a pivotal role in cardiac myocyte development, muscle gene transcription, and hypertrophy. Previously, elevation of intracellular levels of Ca2+ was shown to activate SRF function without involving the Ets family of tertiary complex factors through an unknown regulatory mechanism. Here, we tested the hypothesis that the chromatin remodeling enzymes of class II histone deacetylases (HDAC4) regulate SRF activity in a Ca2+-sensitive manner. Expression of HDAC4 profoundly repressed SRF-mediated transcription in both muscle and nonmuscle cells. Protein interaction studies demonstrated physical association of HDAC4 with SRF in living cells. The SRF/HDAC4 co-association was disrupted by treatment of cells with hypertrophic agonists such as angiotensin-II and a Ca2+ ionophore, ionomycin. Furthermore, activation of Ca2+/calmodulin-dependent protein kinase (CaMK)-IV prevented SRF/HDAC4 interaction and derepressed SRF-dependent transcription activity. The SRF.HDAC4 complex was localized to the cell nucleus, and the activated CaMK-IV disrupted HDAC4/SRF association, leading to export of HDAC4 from the nucleus and stimulation of SRF transcription activity. Thus, these results identify SRF as a functional interacting target of HDAC4 and define a novel tertiary complex factor-independent mechanism for SRF activation by Ca2+/CaMK-mediated signaling.
Collapse
|
49
|
Abstract
Alterations in the differentiated state of vascular smooth muscle cells (SMCs) are known to play a key role in vascular diseases, yet the mechanisms controlling SMC differentiation are still poorly understand. In this review, we discuss our present knowledge of control of SMC differentiation at the transcriptional level, pointing out some common themes, important paradigms, and unresolved issues in SMC-specific gene regulation. We focus primarily on the serum response factor-CArG box-dependent pathway, because it has been shown to play a critical role in regulation of multiple SMC marker genes. However, we also highlight several other important regulatory elements, such as a transforming growth factor beta control element, E-boxes, and MCAT motifs. We present evidence in support of the notion that SMC-specific gene regulation is not controlled by a few SMC-specific transcription factors but rather by complex combinatorial interactions between multiple general and tissue-specific proteins. Finally, we discuss the implications of chromatin remodeling on SMC differentiation.
Collapse
Affiliation(s)
- Meena S Kumar
- Department of Molecular Physiology and Biological Physics, University of Virginia, 415 Lane Rd, MR5 Room 1220, PO Box 801394, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
50
|
Du KL, Ip HS, Li J, Chen M, Dandre F, Yu W, Lu MM, Owens GK, Parmacek MS. Myocardin is a critical serum response factor cofactor in the transcriptional program regulating smooth muscle cell differentiation. Mol Cell Biol 2003; 23:2425-37. [PMID: 12640126 PMCID: PMC150745 DOI: 10.1128/mcb.23.7.2425-2437.2003] [Citation(s) in RCA: 300] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The SAP family transcription factor myocardin functionally synergizes with serum response factor (SRF) and plays an important role in cardiac development. To determine the function of myocardin in the smooth muscle cell (SMC) lineage, we mapped the pattern of myocardin gene expression and examined the molecular mechanisms underlying transcriptional activity of myocardin in SMCs and embryonic stem (ES) cells. The human and murine myocardin genes were expressed in vascular and visceral SMCs at levels equivalent to or exceeding those observed in the heart. During embryonic development, the myocardin gene was expressed abundantly in a precise, developmentally regulated pattern in SMCs. Forced expression of myocardin transactivated multiple SMC-specific transcriptional regulatory elements in non-SMCs. By contrast, myocardin-induced transactivation was not observed in SRF(-/-) ES cells but could be rescued by forced expression of SRF or the SRF DNA-binding domain. Furthermore, expression of a dominant-negative myocardin mutant protein or small-interfering-RNA-induced myocardin knockdown significantly reduced SM22 alpha promoter activity in SMCs. Most importantly, forced expression of myocardin activated expression of the SM22 alpha, smooth muscle alpha-actin, and calponin-h1 genes in undifferentiated mouse ES cells. Taken together, these data demonstrate that myocardin plays an important role in the SRF-dependent transcriptional program that regulates SMC development and differentiation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- COS Cells
- Cell Differentiation/physiology
- Cells, Cultured
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/physiology
- Humans
- Mice
- Microfilament Proteins/genetics
- Molecular Sequence Data
- Muscle Proteins/genetics
- Muscle, Smooth/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocardium/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Organ Specificity
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/physiology
- RNA, Small Interfering/pharmacology
- Rats
- Sequence Alignment
- Serum Response Factor/deficiency
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Stem Cells/cytology
- Stem Cells/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcriptional Activation
Collapse
Affiliation(s)
- Kevin L Du
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|