1
|
Ankyrin Repeat Domain 1 Protein: A Functionally Pleiotropic Protein with Cardiac Biomarker Potential. Int J Mol Sci 2017; 18:ijms18071362. [PMID: 28672880 PMCID: PMC5535855 DOI: 10.3390/ijms18071362] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022] Open
Abstract
The ankyrin repeat domain 1 (ANKRD1) protein is a cardiac-specific stress-response protein that is part of the muscle ankyrin repeat protein family. ANKRD1 is functionally pleiotropic, playing pivotal roles in transcriptional regulation, sarcomere assembly and mechano-sensing in the heart. Importantly, cardiac ANKRD1 has been shown to be highly induced in various cardiomyopathies and in heart failure, although it is still unclear what impact this may have on the pathophysiology of heart failure. This review aims at highlighting the known properties, functions and regulation of ANKRD1, with focus on the underlying mechanisms that may be involved. The current views on the actions of ANKRD1 in cardiovascular disease and its utility as a candidate cardiac biomarker with diagnostic and/or prognostic potential are also discussed. More studies of ANKRD1 are warranted to obtain deeper functional insights into this molecule to allow assessment of its potential clinical applications as a diagnostic or prognostic marker and/or as a possible therapeutic target.
Collapse
|
2
|
Kerkelä R, Ulvila J, Magga J. Natriuretic Peptides in the Regulation of Cardiovascular Physiology and Metabolic Events. J Am Heart Assoc 2015; 4:e002423. [PMID: 26508744 PMCID: PMC4845118 DOI: 10.1161/jaha.115.002423] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Risto Kerkelä
- Department of Pharmacology and Toxicology, Research Unit of Biomedicine, University of Oulu, Finland (R.K., J.U., J.M.) Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Finland (R.K.)
| | - Johanna Ulvila
- Department of Pharmacology and Toxicology, Research Unit of Biomedicine, University of Oulu, Finland (R.K., J.U., J.M.)
| | - Johanna Magga
- Department of Pharmacology and Toxicology, Research Unit of Biomedicine, University of Oulu, Finland (R.K., J.U., J.M.)
| |
Collapse
|
3
|
Matsuoka K, Asano Y, Higo S, Tsukamoto O, Yan Y, Yamazaki S, Matsuzaki T, Kioka H, Kato H, Uno Y, Asakura M, Asanuma H, Minamino T, Aburatani H, Kitakaze M, Komuro I, Takashima S. Noninvasive and quantitative live imaging reveals a potential stress‐responsive enhancer in the failing heart. FASEB J 2014; 28:1870-9. [DOI: 10.1096/fj.13-245522] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ken Matsuoka
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Yoshihiro Asano
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Shuichiro Higo
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Osamu Tsukamoto
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Yi Yan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Satoru Yamazaki
- Department of Cell BiologyNational Cerebral and Cardiovascular Center Research InstituteSuitaJapan
| | - Takashi Matsuzaki
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Hidetaka Kioka
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Hisakazu Kato
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| | - Yoshihiro Uno
- Laboratory of Reproductive EngineeringInstitute of Experimental Animal Sciences, Osaka University Graduate School of MedicineSuitaJapan
| | - Masanori Asakura
- Department of Clinical Research and DevelopmentNational Cerebral and Cardiovascular Center Research InstituteSuitaJapan
| | - Hiroshi Asanuma
- Department of Cardiovascular Science and TechnologyKyoto Prefectural University School of MedicineKyotoJapan
| | - Tetsuo Minamino
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and TechnologyUniversity of TokyoTokyoJapan
| | - Masafumi Kitakaze
- Department of Clinical Research and DevelopmentNational Cerebral and Cardiovascular Center Research InstituteSuitaJapan
| | - Issei Komuro
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Seiji Takashima
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineSuitaJapan
- Department of Medical BiochemistryOsaka University Graduate School of MedicineSuitaJapan
| |
Collapse
|
4
|
Koivisto E, Karkkola L, Majalahti T, Aro J, Tokola H, Kerkelä R, Ruskoaho H. M-CAT element mediates mechanical stretch-activated transcription of B-type natriuretic peptide via ERK activation. Can J Physiol Pharmacol 2011; 89:539-50. [PMID: 21812548 DOI: 10.1139/y11-049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The muscle-CAT (M-CAT) promoter element is found on promoters of most muscle-specific cardiac genes, but its role in cardiac pathology is poorly understood. Here we studied whether the M-CAT element is involved in hypertrophic process activated by mechanical stretch, and identified the intracellular pathways mediating the response. When an in vitro stretch model of cultured neonatal rat cardiomyocytes and luciferase reporter construct driven by rat B-type natriuretic peptide (BNP) promoter were used, mutation of M-CAT element inhibited not only the basal reporter activity (88%), but also the stretch-activated BNP transcription (58%, p < 0.001). Stretch-induced BNP promoter activation was associated with an increase in transcriptional enhancer factor-1 (TEF-1) binding activity after 24 h mechanical stretch (p < 0.05). Inhibition of mitogen-activated protein kinases ERK, JNK, or p38 attenuated stretch-induced BNP activation. Interestingly, as opposed to p38 and JNK, inhibition of ERK had no additional effect on transcriptional activity of BNP promoter harboring the M-CAT mutation, suggesting a pivotal role for ERK in regulating stretch-induced BNP transcription via M-CAT binding site. Finally, immunoprecipitation studies showed that mechanical stretch induced myocyte enhancer factor-2 (MEF-2) binding to TEF-1. These data suggest a central role for M-CAT element in regulation of mechanical stretch-induced hypertrophic response via ERK activation.
Collapse
Affiliation(s)
- Elina Koivisto
- Institute of Biomedicine, Department of Pharmacology and Toxicology, Biocenter Oulu, University of Oulu, Oulu FIN-90014, Finland
| | | | | | | | | | | | | |
Collapse
|
5
|
Abstract
Cardiac-derived peptide hormones were identified more than 25 years ago. An astonishing amount of clinical studies have established cardiac natriuretic peptides and their molecular precursors as useful markers of heart disease. In contrast to the clinical applications, the biogenesis of cardiac peptides has only been elucidated during the last decade. The cellular synthesis including amino acid modifications and proteolytic cleavages has proven considerably more complex than initially perceived. Consequently, the elimination phase of the peptide products in circulation is not yet well characterized. An ongoing characterization of the molecular heterogeneity will help appreciate the biosynthetic capacity of the endocrine heart and could introduce new diagnostic possibilities. Notably, different biosynthetic products may not be equal markers of the same pathophysiological processes. An inefficient post-translational prohormone maturation will also affect the biology of the cardiac natriuretic peptide system. This review aims at summarizing the myocardial synthesis of natriuretic peptides focusing on B-type natriuretic peptide, where new data has disclosed cardiac myocytes as highly competent endocrine cells. The structurally related atrial natriuretic peptide will be mentioned where appropriate, whereas C-type natriuretic peptide will not be considered as a cardiac peptide of relevance in mammalian physiology.
Collapse
|
6
|
Tsika RW, Ma L, Kehat I, Schramm C, Simmer G, Morgan B, Fine DM, Hanft LM, McDonald KS, Molkentin JD, Krenz M, Yang S, Ji J. TEAD-1 overexpression in the mouse heart promotes an age-dependent heart dysfunction. J Biol Chem 2010; 285:13721-35. [PMID: 20194497 DOI: 10.1074/jbc.m109.063057] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
TEA domain transcription factor-1 (TEAD-1) is essential for proper heart development and is implicated in cardiac specific gene expression and the hypertrophic response of primary cardiomyocytes to hormonal and mechanical stimuli, and its activity increases in the pressure-overloaded hypertrophied rat heart. To investigate whether TEAD-1 is an in vivo modulator of cardiac specific gene expression and hypertrophy, we developed transgenic mice expressing hemagglutinin-tagged TEAD-1 under the control of the muscle creatine kinase promoter. We show that a sustained increase in TEAD-1 protein leads to an age-dependent dysfunction. Magnetic resonance imaging revealed decreases in cardiac output, stroke volume, ejection fraction, and fractional shortening. Isolated TEAD-1 hearts revealed decreased left ventricular power output that correlated with increased betaMyHC protein. Histological analysis showed altered alignment of cardiomyocytes, septal wall thickening, and fibrosis, although electrocardiography displayed a left axis shift of mean electrical axis. Transcripts representing most members of the fetal heart gene program remained elevated from fetal to adult life. Western blot analyses revealed decreases in p-phospholamban, SERCA2a, p-CX43, p-GSK-3alpha/beta, nuclear beta-catenin, GATA4, NFATc3/c4, and increased NCX1, nuclear DYKR1A, and Pur alpha/beta protein. TEAD-1 mice did not display cardiac hypertrophy. TEAD-1 mice do not tolerate stress as they die over a 4-day period after surgical induction of pressure overload. These data provide the first in vivo evidence that increased TEAD-1 can induce characteristics of cardiac remodeling associated with cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Richard W Tsika
- Department of Biochemistry, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Ola A, Kerkelä R, Tokola H, Pikkarainen S, Skoumal R, Vuolteenaho O, Ruskoaho H. The mixed-lineage kinase 1-3 signalling pathway regulates stress response in cardiac myocytes via GATA-4 and AP-1 transcription factors. Br J Pharmacol 2010; 159:717-25. [PMID: 20067472 PMCID: PMC2828035 DOI: 10.1111/j.1476-5381.2009.00567.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 09/23/2009] [Accepted: 09/30/2009] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND PURPOSE The mixed-lineage kinases (MLKs) act upstream of mitogen-activated protein kinases, but their role in cardiac biology and pathology is largely unknown. EXPERIMENTAL APPROACH We investigated the effect of a MLK1-3 inhibitor CEP-11004 on G protein-coupled receptor agonist-induced stress response in neonatal rat cardiac myocytes in culture. KEY RESULTS CEP-11004 administration dose-dependently attenuated phenylephrine and endothelin-1 (ET-1)-induced c-Jun N-terminal kinase activation. MLK inhibition also reduced ET-1- and phenylephrine-induced phosphorylation of p38 mitogen-activated protein kinase. In contrast, phenylephrine-induced extracellular signal-regulated kinase phosphorylation was further up-regulated by CEP-11004. ET-1 increased activator protein-1 binding activity 3.5-fold and GATA-binding protein 4 (GATA-4) binding activity 1.8-fold, both of which were attenuated with CEP-11004 administration by 59% and 63% respectively. Phenylephrine induced activator protein-1 binding activity by 2.6-fold, which was decreased by 81% with CEP-11004 administration. Phenylephrine also induced a 3.7-fold increase in the transcriptional activity of B-type natriuretic peptide (BNP), which was attenuated by 41% with CEP-11004 administration. In agreement, MLK inhibition also reduced hypertrophic agonist-induced secretion of immunoreactive atrial natriuretic peptide and BNP. CONCLUSIONS AND IMPLICATIONS These results showed that inhibition of the MLK1-3 signalling pathway was sufficient for suppressing the activity of key nuclear effectors (GATA-4 and activator protein-1 transcription factors) in cardiac hypertrophy, and attenuated the agonist-induced atrial natriuretic peptide secretion and activation of BNP gene transcription.
Collapse
Affiliation(s)
- A Ola
- Institute of Biomedicine, Department of Pharmacology and Toxicology, Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
Atrial and brain natriuretic peptides (ANP and BNP, respectively) are cardiac hormones. During cardiac development, their expression is a maker of cardiomyocyte differentiation and is under tight spatiotemporal regulation. After birth, however, their ventricular expression is only up-regulated in response to various cardiovascular diseases. As a result, analysis of ANP and BNP gene expression has led to discoveries of transcriptional regulators and signaling pathways involved in both cardiac differentiation and cardiac disease. Studies using genetically engineered mice have shed light on the molecular mechanisms regulating ANP and BNP gene expression, as well as the physiological and pathophysiological relevance of the cardiac natriuretic peptide system. In this review we will summarize what is currently known about their regulation and the significance of ANP and BNP as hormones derived from the heart.
Collapse
Affiliation(s)
- Koichiro Kuwahara
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan.
| | | |
Collapse
|
9
|
McWhinney C, Robishaw JD. Myocyte-specific M-CAT and MEF-1 elements regulate G-protein gamma 3 gene (gamma3) expression in cardiac myocytes. DNA Cell Biol 2008; 27:367-76. [PMID: 18613791 DOI: 10.1089/dna.2008.0754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Little is known regarding the mechanisms that control the expression of G-protein alpha, beta, and gamma subtypes. We have previously shown that the G-protein gamma(3) gene is expressed in the heart, brain, lung, spleen, kidney, muscle, and testis in mice. We have also reported that the G-protein gamma(3) subunit is expressed in rat cardiac myocytes, but not in cardiac fibroblasts. Other studies have shown that the gamma(3) subunit couples to the angiotensin A1A receptor in portal vein myocytes, and has been shown to mediate beta-adrenergic desensitization in cardiac myocytes treated with atorvastatin. In the present study, we evaluated G-protein gamma(3) promoter-luciferase reporter constructs in primary myocytes to identify key regulatory promoter regions. We identified two important regions of the promoter (upstream promoter region [UPR] and downstream promoter region [DPR]), which are required for expression in cardiac myocytes. We observed that removal of 48 bp in the UPR diminished gene transcription by 75%, and that the UPR contains consensus elements for myocyte-specific M-CAT and myocyte enhancer factor 1 (MEF-1) elements. The UPR and DPR share transcription factor elements for myocyte-specific M-CAT element. We observed that cardiac myocyte proteins bind to gamma(3) oligonucleotides containing transcription factor elements for myocyte-specific M-CAT and MEF-1. Myocyte-specific M-CAT proteins were supershifted with transcriptional enhancer factor-1 (TEF-1) antibodies binding to the gamma(3) M-CAT element, which is in agreement with reports showing that the M-CAT element binds the TEF-1 family of transcription factors. The 150 bp DPR contains three M-CAT elements, an INR element, an upstream stimulatory factor 1 element, and the transcription start site. We have shown that myocyte gamma(3) gene expression is regulated by myocyte-specific M-CAT and MEF-1 elements.
Collapse
Affiliation(s)
- Charlene McWhinney
- Biomedical Sciences Department, New York College of Osteopathic Medicine, Old Westbury, New York 11568-8000, USA.
| | | |
Collapse
|
10
|
Affiliation(s)
- David G Gardner
- Diabetes Center, University of California at San Francisco, San Francisco, CA 94143-0540, USA.
| | | | | | | |
Collapse
|
11
|
Raizada A, Bhandari S, Khan MA, Singh HV, Thomas S, Sarabhai V, Singh N, Trehan N. Brain type natriuretic peptide (BNP)-A marker of new millennium in diagnosis of congestive heart failure. Indian J Clin Biochem 2007; 22:4-9. [PMID: 23105644 PMCID: PMC3454260 DOI: 10.1007/bf02912873] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The burden of disease in patients with congestive heart failure is high. The future of BNP looks promising as it may be a better diagnostic tool for the diagnosis of CHF in developing countries in new millennium. Natriuretic peptide hormones, a family of vasoactive peptides with many favourable physiological properties, have emerged as important contenders for development of diagnostic tools and therapeutic agents in cardiovascular disease. Measurement of B-type natriuretic peptide has become as an easy-to-perform bedside test. The clinical and diagnostic significance of the measurement of plasma Nt-proBNP in the diseases of the cardiovascular system with particular emphasis on the assessment of patients with heart failure and their effects on predicting survival rate. The plasma levels of Nt-proBrain Natriuretic peptide responds more vigorously after myocardial infarction than those of other natriuretic peptides. This article is an attempt to give a short overview on the utility of BNP-blood levels for the diagnosis and treatment of heart failure.
Collapse
Affiliation(s)
- Arun Raizada
- Department of Clinical Biochemistry and Cardiology, Escorts Heart Insitute & Research Centre, 110025 New Delhi
| | - Suman Bhandari
- Department of Clinical Biochemistry and Cardiology, Escorts Heart Insitute & Research Centre, 110025 New Delhi
| | - Muzaiyan Ahmed Khan
- Department of Biochemistry, People's College of Dental Sciences & Research Centre, 462018 Bhopal
| | | | - Sherin Thomas
- Department of Clinical Biochemistry and Cardiology, Escorts Heart Insitute & Research Centre, 110025 New Delhi
| | - Vikram Sarabhai
- Department of Clinical Biochemistry and Cardiology, Escorts Heart Insitute & Research Centre, 110025 New Delhi
| | - Neelima Singh
- Department of Biochemistry, G. R. Medical College, 474002 Gwalior
| | - Naresh Trehan
- Department of Clinical Biochemistry and Cardiology, Escorts Heart Insitute & Research Centre, 110025 New Delhi
| |
Collapse
|
12
|
Tenhunen O, Szokodi I, Ruskoaho H. Posttranscriptional activation of BNP gene expression in response to increased left ventricular wall stress: role of calcineurin and PKC. ACTA ACUST UNITED AC 2005; 128:187-96. [PMID: 15837527 DOI: 10.1016/j.regpep.2004.12.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To study the molecular mechanisms for load-induced activation of BNP gene expression, increased wall stress was imposed on isolated isovolumetrically beating adult rat hearts by distension of a fluid filled balloon within the left ventricle. The wall stress for 2 h resulted in a 1.6-fold increase in the expression of BNP gene and a 2.0-fold increase of the c-fos gene. The inhibition of transcription by actinomycin D significantly decreased the baseline BNP mRNA levels but the wall stretch-induced increase of the gene expression remained unaffected. In contrast, the protein synthesis inhibitor cycloheximide increased baseline BNP mRNA levels and abolished the load-induced activation of gene expression. Furthermore, we studied the effects of protein kinase C (PKC), calcineurin and protein phosphatase 2A (PP2A) inhibition to characterize the role of intracellular pathways in the stretch-induced gene expression in the left ventricle. The expression of BNP and c-fos genes were not influenced by calcineurin, PP2A and PKC inhibition. In conclusion, we showed that the stretch-induced activation of BNP gene expression by increased left ventricular wall stress is independent of transcriptional mechanisms and dependent on protein synthesis. Moreover, our results suggest that the load-induced activation of BNP gene expression is independent of calcineurin, PKC and PP2A.
Collapse
Affiliation(s)
- Olli Tenhunen
- Department of Pharmacology and Toxicology, Faculty of Medicine, Biocenter Oulu, University of Oulu, Finland
| | | | | |
Collapse
|
13
|
Abstract
After brain natriuretic peptide (BNP) was isolated in 1988, rapid progress was made in cloning its cDNA and gene, facilitating studies of tissue-specific expression and molecular regulation of gene expression. This review focuses on the molecular determinants of regulation of the rat and human BNP genes, including signaling pathways that impact on changes in gene expression and cis regulatory elements responsive to these signaling pathways. For both rat and human genes, elements in the proximal promoter (-124 to -80), including GATA, MCAT, and AP-1-like, have been shown to contribute to basal and inducible regulation. More distal elements in the human BNP gene respond to calcium signals (an NF-AT site at -927), thyroid hormone (a thyroid-responsive element at -1000), and mechanical stretch (shear stress-responsive elements at -652 and -162). Understanding how BNP is regulated by signaling molecules that are activated in the hypertrophied and ischemic heart should be useful in understanding the underlying pathology. This may lead to therapeutic strategies that prevent hypertrophy while allowing for the beneficial effects of BNP production.
Collapse
Affiliation(s)
- Margot C LaPointe
- Hypertension and Vascular Research Division, Department of Medicine, Henry Ford Hospital, 2799 W. Grand Blvd., Detroit, MI 48202, USA.
| |
Collapse
|
14
|
Ma KK, Banas K, de Bold AJ. Determinants of inducible brain natriuretic peptide promoter activity. ACTA ACUST UNITED AC 2005; 128:169-76. [PMID: 15837525 DOI: 10.1016/j.regpep.2004.12.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Atrial natriuretic factor (ANF) and brain natriuretic peptide (BNP) are polypeptide hormones belonging to the cardiac-derived mammalian natriuretic peptide system. These hormones share the same biological properties and receptors and both play important roles in the maintenance of fluid and electrolyte balance and in cardiovascular growth. Most hemodynamic and neurohumoral stimuli can coordinately increase ANF and BNP gene expression. However, instances of discoordinated ANF and BNP gene expression have been described, providing an opportunity for investigating the mechanisms that differentially regulate the expression of the natriuretic peptide genes. For example, exposure of cardiocytes in culture to certain pro-inflammatory cytokines and conditioned medium from mixed lymphocyte cultures upregulate BNP but not ANF gene expression. BNP promoter activity is also upregulated under these conditions but the cis-acting elements involved in this phenomenon are not known. In comparison to the ANF gene, less is known about BNP promoter consensus elements that regulate gene expression by mechanical or neurohumoral agonists. A number of cis-acting elements for GATA, Nkx2.5, NF-kappaB and TEF transcription factors have recently been identified within the BNP promoter that regulate BNP expression in response to specific agonists. This review focuses on the information available regarding cis-acting determinants responsible for inducible BNP transcription.
Collapse
Affiliation(s)
- Kenneth K Ma
- Cardiovascular Endocrinology Laboratory, University of Ottawa Heart Institute, Department of Cellular and Molecular Medicine, Faculty of Medicine, Canada
| | | | | |
Collapse
|
15
|
Pikkarainen S, Tokola H, Majalahti-Palviainen T, Kerkela R, Hautala N, Bhalla SS, Charron F, Nemer M, Vuolteenaho O, Ruskoaho H. GATA-4 is a nuclear mediator of mechanical stretch-activated hypertrophic program. J Biol Chem 2003; 278:23807-16. [PMID: 12704188 DOI: 10.1074/jbc.m302719200] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In overloaded heart the cardiomyocytes adapt to increased mechanical and neurohumoral stress by activation of hypertrophic program, resulting in morphological changes of individual cells and specific changes in gene expression. Accumulating evidence suggests an important role for the zinc finger transcription factor GATA-4 in hypertrophic agonist-induced cardiac hypertrophy. However, its role in stretch-induced cardiomyocyte hypertrophy is not known. We employed an in vitro mechanical stretch model of cultured cardiomyocytes and used rat B-type natriuretic peptide promoter as stretch-sensitive reporter gene. Stretch transiently increased GATA-4 DNA binding activity and transcript levels, which was followed by increases in the expression of B-type natriuretic peptide as well as atrial natriuretic peptide and skeletal alpha-actin genes. The stretch inducibility mapped primarily to the proximal 520 bp of the B-type natriuretic peptide promoter. Mutational studies showed that the tandem GATA consensus sites of the proximal promoter in combination with an Nkx-2.5 binding element are critical for stretch-activated B-type natriuretic peptide transcription. Inhibition of GATA-4 protein production by adenovirus-mediated transfer of GATA-4 antisense cDNA blocked stretch-induced increases in B-type natriuretic peptide transcript levels and the sarcomere reorganization. The proportion of myocytes with assembled sarcomeres in control adenovirus-infected cultures increased from 14 to 59% in response to stretch, whereas the values for GATA-4 antisense-treated cells were 6 and 13%, respectively. These results show that activation of GATA-4, in cooperation with a factor binding on Nkx-2.5 binding element, is essential for mechanical stretch-induced cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Sampsa Pikkarainen
- Department of Pharmacology, Biocenter Oulu, University of Oulu, PO Box 5000, FIN-90014 University of Oulu, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Clerk A, Cole SM, Cullingford TE, Harrison JG, Jormakka M, Valks DM. Regulation of cardiac myocyte cell death. Pharmacol Ther 2003; 97:223-61. [PMID: 12576135 DOI: 10.1016/s0163-7258(02)00339-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiac myocyte death, whether through necrotic or apoptotic mechanisms, is a contributing factor to many cardiac pathologies. Although necrosis and apoptosis are the widely accepted forms of cell death, they may utilize the same cell death machinery. The environment within the cell probably dictates the final outcome, producing a spectrum of response between the two extremes. This review examines the probable mechanisms involved in myocyte death. Caspases, the generally accepted executioners of apoptosis, are significant in executing cardiac myocyte death, but other proteases (e.g., calpains, cathepsins) also promote cell death, and these are discussed. The two principal cell death pathways (death receptor- and mitochondrial-mediated) are described in relation to the emerging structural information for the principal proteins, and they are discussed relative to current understanding of myocyte cell death mechanisms. Whereas the mitochondrial pathway is probably a significant factor in myocyte death in both acute and chronic phases of myocardial diseases, the death receptor pathway may prove significant in the longer term. The Bcl-2 family of proteins are key regulators of the mitochondrial death pathway. These proteins are described and their possible functions are discussed. The commitment to cell death is also influenced by protein kinase cascades that are activated in the cell. Whereas certain pathways are cytoprotective (e.g., phosphatidylinositol 3'-kinase), the roles of other kinases are less clear. Since myocyte death is implicated in a number of cardiac pathologies, attenuation of the death pathways may prove important in ameliorating such disease states, and possible therapeutic strategies are explored.
Collapse
Affiliation(s)
- Angela Clerk
- NHLI Division (Cardiac Medicine Section), Faculty of Medicine, Imperial College of Science, Technology and Medicine, Flower's Building, Armstrong Road, South Kensington, London SW7 2AZ, UK.
| | | | | | | | | | | |
Collapse
|
17
|
Pikkarainen S, Tokola H, Kerkelä R, Majalahti-Palviainen T, Vuolteenaho O, Ruskoaho H. Endothelin-1-specific activation of B-type natriuretic peptide gene via p38 mitogen-activated protein kinase and nuclear ETS factors. J Biol Chem 2003; 278:3969-75. [PMID: 12446726 DOI: 10.1074/jbc.m205616200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Terminally differentiated cardiac myocytes adapt to mechanical and neurohumoral stress via morphological changes of individual cells accompanied by reactivation of fetal pattern of gene expression. Endothelin-1, a powerful paracrine mediator of myocyte growth, induces similar changes in cultured cardiac myocytes as those seen in hypertrophied heart in vivo. By using rat B-type natriuretic peptide promoter, we identified a novel ETS binding sequence, on which nuclear protein binding is activated in endothelin-1-treated cultured cardiac myocytes. This sequence binds ETS-like gene-1 transcription factor and mediates endothelin-1-specific activation of transcription, but not responses to increased calcium signaling via l-type calcium channels, angiotensin II treatment, or mechanical stretch of myocytes. Interestingly, endothelin-1 activated signaling converges via p38 mitogen-activated protein kinase-dependent mechanism on ETS binding site, whereas this element inhibits extracellular signal-regulated kinase activated transcription. In conclusion, given the fundamental role of the interaction of mitogen-activated protein kinases and ETS factors in regulation of eukaryotic cell differentiation, growth, and oncogenesis, these results provide the unique evidence of a endothelin-1- and mitogen-activated protein kinase-regulated ETS factor pathway for cardiac myocytes.
Collapse
Affiliation(s)
- Sampsa Pikkarainen
- Department of Pharmacology and Toxicology, Biocenter Oulu, University of Oulu, P. O. Box 5000, Finland
| | | | | | | | | | | |
Collapse
|
18
|
Affiliation(s)
- Kirkwood F Adams
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7075, USA.
| | | | | |
Collapse
|
19
|
Maeda T, Maeda M, Stewart AFR. TEF-1 transcription factors regulate activity of the mouse mammary tumor virus LTR. Biochem Biophys Res Commun 2002; 296:1279-85. [PMID: 12207913 DOI: 10.1016/s0006-291x(02)02085-5] [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] [Indexed: 01/19/2023]
Abstract
The mouse mammary tumor virus long terminal repeat (LTR) is a potent transcriptional enhancer. We identified several putative binding sites for the TEF-1 family of transcription factors (TEF-1, RTEF-1, DTEF-1, and ETF) in the proximal negative regulatory element of the LTR. Gel mobility shift assays revealed strong TEF-1 factor binding to one site using nuclear extracts from CV-1 cells and from the human breast cancer cell line MCF-7. Mutation of this site increased basal activity of the LTR. In transient transfection assays, TEF-1 squelched the basal LTR activity and completely abrogated the response to the glucocorticoid dexamethasone. RTEF-1 and DTEF-1 had little effect on the basal activity, whereas ETF activated the LTR. These TEF-1 factors also interfered with the response to dexamethasone. Taken together, our results reveal an important new role for TEF-1 factors in regulating MMTV LTR activity and suggest that TEF-1 factors might participate in mammary tumorigenesis.
Collapse
Affiliation(s)
- Tomoji Maeda
- Cardiovascular Institute, School of Medicine, University of Pittsburgh, BST 1704.3, 200 Lothrop Street, 15213, Pittsburgh, PA, USA
| | | | | |
Collapse
|
20
|
Maeda T, Sepulveda J, Chen HH, Stewart AFR. Alpha(1)-adrenergic activation of the cardiac ankyrin repeat protein gene in cardiac myocytes. Gene 2002; 297:1-9. [PMID: 12384280 DOI: 10.1016/s0378-1119(02)00924-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiac ankyrin repeat protein (CARP) is a nuclear transcription cofactor that is activated by multiple signaling pathways in hypertrophic cardiac myocytes. Since CARP has been reported to be a transcriptional co-repressor, its activation during hypertrophy might contribute to the deregulation of gene expression leading to heart failure. Here, we found that alpha(1)-adrenergic signaling activates CARP mRNA expression in rat cardiac myocytes. To examine how alpha(1)-adrenergic signaling activates the CARP gene, a 660 bp fragment of the mouse CARP promoter was cloned. Previous reports suggested that the mouse CARP promoter was dependent on the GATA4 transcription factor whereas the human CARP promoter was dependent on transcriptional enhancer factor-1 (TEF-1). TEF-1 and GATA4 transcription factors, known mediators of alpha(1)-adrenergic signaling, bound to the mouse CARP promoter at several sites as determined by gel mobility shift assays. These sites are highly conserved between the mouse and human promoters, suggesting that they are functionally important in both. Mutation analysis showed that binding of TEF-1 factors is required for basal activity of the CARP promoter in cardiac myocytes. However, over-expression of TEF-1 factors could not potentiate the response of the CARP promoter to alpha(1)-adrenergic stimulation. On the other hand, the alpha(1)-adrenergic response was potentiated by GATA4 over-expression. Taken together, our results demonstrate that alpha(1)-adrenergic signaling regulates CARP expression in cardiac myocytes, in part through the transcription factor GATA4.
Collapse
Affiliation(s)
- Tomoji Maeda
- Cardiovascular Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
21
|
Maeda T, Mazzulli JR, Farrance IKG, Stewart AFR. Mouse DTEF-1 (ETFR-1, TEF-5) is a transcriptional activator in alpha 1-adrenergic agonist-stimulated cardiac myocytes. J Biol Chem 2002; 277:24346-52. [PMID: 11986313 DOI: 10.1074/jbc.m201171200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
alpha(1)-Adrenergic signaling in cardiac myocytes activates the skeletal muscle alpha-actin gene through an MCAT cis-element, the binding site of the transcriptional enhancer factor-1 (TEF-1) family of transcription factors. TEF-1 accounts for more than 85% of the MCAT binding activity in neonatal rat cardiac myocytes. Other TEF-1 family members account for the rest. Although TEF-1 itself has little effect on the alpha(1)-adrenergic activation of skeletal muscle alpha-actin, the related factor RTEF-1 augments the response and is a target of alpha(1)-adrenergic signaling. Here, we examined another TEF-1 family member expressed in cardiac muscle, DTEF-1, and observed that it also augmented the alpha(1)-adrenergic response of skeletal muscle alpha-actin. A DTEF-1 peptide-specific antibody revealed that endogenous DTEF-1 accounts for up to 5% of the MCAT binding activity in neonatal rat cardiac myocytes. A TEF-1/DTEF-1 chimera suggests that alpha(1)-adrenergic signaling modulates DTEF-1 function. Orthophosphate labeling and immunoprecipitation of an epitope-tagged DTEF-1 showed that DTEF-1 is phosphorylated in vivo. alpha(1)-Adrenergic stimulation increased while phosphatase treatment lowered the MCAT binding by DTEF-1 and the endogenous non-TEF-1 MCAT-binding factor. In contrast, alpha(1)-adrenergic stimulation did not alter, and phosphatase treatment increased, MCAT binding of TEF-1 and RTEF-1. Taken together, these results suggest that DTEF-1 is a target for alpha(1)-adrenergic activation of the skeletal muscle alpha-actin gene in neonatal rat cardiac myocytes.
Collapse
Affiliation(s)
- Tomoji Maeda
- Cardiovascular Institute, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
22
|
Abstract
Brain natriuretic peptide (BNP) is a cardiac hormone constitutively expressed in the adult heart. We previously showed that the human BNP (hBNP) proximal promoter region from -127 to -40 confers myocyte-specific expression. The proximal hBNP promoter contains several putative cis elements. Here we tested whether the proximal GATA element plays a role in basal and inducible regulation of the hBNP promoter. The hBNP promoter was coupled to a luciferase reporter gene (1818hBNPLuc) and transferred into neonatal ventricular myocytes (NVM), and luciferase activity was measured as an index of hBNP promoter activity. Mutation of the putative GATA element at -85 of the hBNP promoter [1818(mGATA)hBNPLuc] reduced activity by 97%. To study transactivation of the hBNP promoter, we co-transfected 1818hBNPLuc with the GATA-4 expression vector. GATA-4 activated 1818hBNPLuc, and this effect was eliminated by mutation of the proximal GATA element. Electrophoretic mobility shift assay showed that an oligonucleotide containing the hBNP GATA motif bound to cardiomyocyte nuclear protein, which was competed for by a consensus GATA oligonucleotide but not a mutated hBNP GATA element. The beta-adrenergic agonist isoproterenol and its second messenger cAMP stimulated hBNP promoter activity and binding of nuclear protein to the proximal GATA element. Thus the GATA element in the proximal hBNP promoter is involved in both basal and inducible transcriptional regulation in cardiac myocytes.
Collapse
Affiliation(s)
- Quan He
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
23
|
Tse MY, Watson JD, Sarda IR, Flynn TG, Pang SC. Expression of B-type natriuretic peptide in atrial natriuretic peptide gene disrupted mice. Mol Cell Biochem 2001; 219:99-105. [PMID: 11354260 DOI: 10.1023/a:1011087513774] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) are two hormones produced and secreted by the heart to control blood pressure, body fluid homeostasis and electrolyte balance. Each peptide binds to a common family of 3 receptors (GC-A, GC-B and C-receptor) with varying degrees of affinity. The proANP gene disrupted mouse model provides an excellent opportunity to examine the regulation and expression of BNP in the absence ofANP. A new radioimmunoassay (RIA) was developed in order to measure mouse BNP peptide levels in the plasma, atrium and ventricle of the mouse. A detection limit of 3-6 pg/tube was achieved by this assay. Results show that plasma and ventricular level of BNP were unchanged among the three genotypes of mice. However, a significant decrease in the BNP level was noted in the atrium. The homozygous mutant (ANP-/-) had undetectable levels of BNP in the atrium, while the heterozygous (ANP+/-) and wild-type (ANP+/+) mice had 430 and 910 pg/mg in the atrium, respectively. Northern Blot analysis shows the ANP-/- mice has a 40% reduction of BNP mRNA level in the atrium and a 5-fold increase in the ventricle as compared with that of the ANP+/+ mouse. Our data suggest that there is a compensatory response of BNP expression to proANP gene disruption. Despite the changes in the atrial and ventricular tissue mRNA and peptide levels, the plasma BNP level remains unaltered in the ANP-/- mice. We conclude that the inability of BNP to completely compensate for the lack of ANP eventually leads to chronic hypertension in the proANP gene disrupted mice.
Collapse
Affiliation(s)
- M Y Tse
- Department of Anatomy and Cell Biology, Queen 's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
24
|
Tokola H, Hautala N, Marttila M, Magga J, Pikkarainen S, Kerkelä R, Vuolteenaho O, Ruskoaho H. Mechanical load-induced alterations in B-type natriuretic peptide gene expression. Can J Physiol Pharmacol 2001. [DOI: 10.1139/y01-031] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), and C-type natriuretic peptide are the known members of the mammalian natriuretic peptide system. Like ANP, BNP is a natriuretic and diuretic hormone that also causes peripheral vasodilation and inhibition of the sympathetic and renin-angiotensin systems. Although originally isolated from porcine brain, the BNP gene is expressed in a specific manner in cardiac myocytes in both the atria and the ventricles, but it is mainly released from the ventricles. The major determinant of BNP secretion is wall stretch, and the levels of BNP mRNA increase substantially in response to cardiac overload. In the clinical setting, BNP appears to be the most powerful neurohumoral predictor of left-ventricular function and prognosis. An acute increase in BNP gene expression occurs within 1 h and mimics the rapid induction of proto-oncogenes in response to hemodynamic stress. BNP can be used as a myocyte-specific marker to identify mechanisms that couple acute mechanical overload to alterations in cardiac gene expression. This paper is focused on the mechanisms that regulate BNP gene expression in cardiac overload. Particularly, autocrine-paracrine factors as well as cytoplasmic signaling pathways and transcription factors involved in mechanical stretch-induced BNP gene expression are discussed.Key words: gene expression, mechanical load, natriuretic peptides, paracrine factors, transcription factors.
Collapse
|
25
|
O'Connell TD, Rokosh DG, Simpson PC. Cloning and characterization of the mouse alpha1C/A-adrenergic receptor gene and analysis of an alpha1C promoter in cardiac myocytes: role of an MCAT element that binds transcriptional enhancer factor-1 (TEF-1). Mol Pharmacol 2001; 59:1225-34. [PMID: 11306707 DOI: 10.1124/mol.59.5.1225] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
alpha1-Adrenergic receptor (AR) subtypes in the heart are expressed by myocytes but not by fibroblasts, a feature that distinguishes alpha1-ARs from beta-ARs. Here we studied myocyte-specific expression of alpha1-ARs, focusing on the subtype alpha1C (also called alpha1A), a subtype implicated in cardiac hypertrophic signaling in rat models. We first cloned the mouse alpha1C-AR gene, which consisted of two exons with an 18 kb intron, similar to the alpha1B-AR gene. The receptor coding sequence was >90% homologous to that of rat and human. alpha1C-AR transcription in mouse heart was initiated from a single Inr consensus sequence at -588 from the ATG; this and a putative polyadenylation sequence 8.5 kb 3' could account for the predominant 11 kb alpha1C mRNA in mouse heart. A 5'-nontranscribed fragment of 4.4 kb was active as a promoter in cardiac myocytes but not in fibroblasts. Promoter activity in myocytes required a single muscle CAT (MCAT) element, and this MCAT bound in vitro to recombinant and endogenous transcriptional enhancer factor-1. Thus, alpha1C-AR transcription in cardiac myocytes shares MCAT dependence with other cardiac-specific genes, including the alpha- and beta-myosin heavy chains, skeletal alpha-actin, and brain natriuretic peptide. However, the mouse alpha1C gene was not transcribed in the neonatal heart and was not activated by alpha1-AR and other hypertrophic agonists in rat myocytes, and thus differed from other MCAT-dependent genes and the rat alpha1C gene.
Collapse
Affiliation(s)
- T D O'Connell
- Cardiology Division and Research Service, Veterans Affairs Medical Center, San Francisco, California, USA
| | | | | |
Collapse
|
26
|
Silberbach M, Roberts CT. Natriuretic peptide signalling: molecular and cellular pathways to growth regulation. Cell Signal 2001; 13:221-31. [PMID: 11306239 DOI: 10.1016/s0898-6568(01)00139-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The natriuretic peptides (NPs) constitute a family of polypeptide hormones that regulate mammalian blood volume and blood pressure. The ability of the NPs to modulate cardiac hypertrophy and cell proliferation as well is now beginning to be recognized. The NPs interact with three membrane-bound receptors, all of which contain a well-characterized extracellular ligand-binding domain. The R1 subclass of NP receptors (NPR-A and NPR-B) contains a C-terminal guanylyl cyclase domain and is responsible for most of the NPs downstream actions through their ability to generate cGMP. The R2 subclass lacks an obvious catalytic domain and functions primarily as a clearance receptor. This review focuses on the signal transduction pathways initiated by ligand binding and other factors that help to determine signalling specificities, including allosteric factors modulating cGMP generation, receptor desensitization, the activation and function of cGMP-dependent protein kinase (PKG), and identification of potential nuclear or cytoplasmic targets such as the mitogen-activated protein kinase signalling (MAPK) cascade. The inhibition of cardiac growth and hypertrophy may be an important but underappreciated action of the NP signalling system.
Collapse
Affiliation(s)
- M Silberbach
- Division of Pediatric Cardiology, Department of Pediatrics, Doernbecher Children's Hospital, UHN-60, 3181 SW Sam Jackson Park Road, 97201, Portland, OR, USA.
| | | |
Collapse
|
27
|
He Q, Wang D, Yang XP, Carretero OA, LaPointe MC. Inducible regulation of human brain natriuretic peptide promoter in transgenic mice. Am J Physiol Heart Circ Physiol 2001; 280:H368-76. [PMID: 11123253 DOI: 10.1152/ajpheart.2001.280.1.h368] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Studies have shown that brain natriuretic peptide (BNP) gene expression is rapidly induced in the infarcted heart and that plasma BNP levels reflect the degree of left ventricular dysfunction. Our previous in vitro work using transiently transfected neonatal rat cardiac myocytes has shown that the human BNP (hBNP) promoter, in particular a region extending from -127 to -40 relative to the start site of transcription, is more active in cardiac myocytes than in fibroblasts. To study tissue-specific and transcriptional regulation of the hBNP gene in vivo, we generated transgenic mice containing the proximal hBNP promoter (-408 to +100) coupled to a luciferase reporter gene. In four lines of transgenic mice, luciferase activity was approximately 33- to 100-fold higher in the heart than in other tissues, including the whole brain. To test whether the transgene responded to a pathophysiological stimulus, we induced infarction by coronary artery ligation. Luciferase activity was fivefold higher in the infarcted region of the left ventricle at 48 h than in sham-operated animals and remained elevated for 4 wk. Endogenous BNP mRNA was similarly increased in the infarcted hearts of a separate group of mice. We conclude that 1) the proximal 408-bp region of the hBNP promoter confers cardiac-specific expression and 2) myocardial infarction activates the proximal hBNP promoter in vivo. These data suggest that we have a valid model for the study of basal and inducible regulation of the hBNP gene in vivo.
Collapse
Affiliation(s)
- Q He
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | |
Collapse
|
28
|
Kodama H, Fukuda K, Pan J, Sano M, Takahashi T, Kato T, Makino S, Manabe T, Murata M, Ogawa S. Significance of ERK cascade compared with JAK/STAT and PI3-K pathway in gp130-mediated cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2000; 279:H1635-44. [PMID: 11009450 DOI: 10.1152/ajpheart.2000.279.4.h1635] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We compared the role of the Raf-1/mitogen-activated protein kinase/extracellular signal-regulated protein kinase (MEK)/extracellular signal-regulated protein kinase (ERK)/p90(RSK) cascade in gp130-mediated cardiac hypertrophy with the contribution of the Janus kinase (JAK)/signal transduction and activation of transcription (STAT) and phosphatidylinositide 3-kinase (PI3-K) pathways. Primary cultured neonatal rat cardiomyocytes were stimulated with leukemia inhibitory factor (LIF). LIF sequentially activated Raf-1, MEK1/2, ERK1/2, and p90(RSK). We used PD-98059 (a specific MEK inhibitor), AG-490 (a JAK2 inhibitor), and wortmannin (a PI3-K inhibitor) to confirm that this cascade was independent of the JAK/STAT and PI3-K/p70 S6 kinase (S6K) pathways. PD-98059, AG-490, and wortmannin suppressed the LIF-induced increase in [(3)H]phenylalanine uptake by 54.7, 21.5, and 25.6%, respectively, and inhibited the increase in cell area by 61.2, 42.8, and 39.2%, respectively. Reorganization of myofilaments was predominantly suppressed by AG-490. LIF-induced expression of c-fos, brain natriuretic peptide, and skeletal alpha-actin mRNA was markedly suppressed by PD-98059 and moderately suppressed by wortmannin and AG-490. Atrial natriuretic peptide was significantly suppressed by AG-490. These findings indicate that this pathway is critically involved in protein synthesis, induction of c-fos, brain natriuretic peptide, and skeletal alpha-actin expression and is partially involved in myofilament reorganization and atrial natriuretic peptide induction in gp130-mediated cardiac hypertrophy.
Collapse
Affiliation(s)
- H Kodama
- Cardiopulmonary Division, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pemberton CJ, Johnson ML, Yandle TG, Espiner EA. Deconvolution analysis of cardiac natriuretic peptides during acute volume overload. Hypertension 2000; 36:355-9. [PMID: 10988264 DOI: 10.1161/01.hyp.36.3.355] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiac natriuretic peptides, especially amino terminal pro-Brain Natriuretic Peptide (NT-proBNP), are emerging as powerful circulating markers of cardiac function. However, the in vivo secretion and elimination (t1/2) of these peptides during acute volume overload have not been studied. We present the first report of the secretion and elimination of cardiac natriuretic peptides, based on deconvolution analysis of endogenous ovine plasma levels measured by specific radioimmunoassay. Four normal, conscious sheep underwent rapid right ventricular pacing (225 bpm) for 1 hour to stimulate acute cardiac natriuretic peptide release. Plasma samples and right atrial pressure measurements were taken at regular intervals 30 minutes before, during, and 4 hours after pacing. Baseline right atrial pressure significantly increased (P:=0.02) during the 1 hour of pacing in association with a prompt increase in plasma BNP (P:=0.03), atrial natriuretic peptide (P:=0.01), and NT-proBNP (P:=0.02). Deconvolution analysis showed that the t1/2 of NT-proBNP (69.6+/-10.8 minutes) was 15-fold longer than BNP (4.8+/-1. 0 minutes). Despite sustained increases in atrial pressure, cardiac secretion of natriuretic peptides (particularly atrial natriuretic peptide) fell during the pacing period, suggesting a finite source of peptide for secretion. Size-exclusion high-performance liquid chromatography revealed NT-proBNP to be a single immunoreactive peak, whereas BNP comprised at least 2 immunoreactive forms. These findings, especially the prompt secretion of BNP and the prolonged t1/2 of NT-proBNP, clarify the metabolism of BNP forms and help to explain the diagnostic value of NT-proBNP measurement as a sensitive marker of ventricular function.
Collapse
Affiliation(s)
- C J Pemberton
- Christchurch Cardioendocrine Research Group, Christchurch School of Medicine, University of Otago, and Christchurch Hospital, Christchurch, New Zealand
| | | | | | | |
Collapse
|
30
|
Aihara Y, Kurabayashi M, Tanaka T, Takeda SI, Tomaru K, Sekiguchi KI, Ohyama Y, Nagai R. Doxorubicin represses CARP gene transcription through the generation of oxidative stress in neonatal rat cardiac myocytes: possible role of serine/threonine kinase-dependent pathways. J Mol Cell Cardiol 2000; 32:1401-14. [PMID: 10900167 DOI: 10.1006/jmcc.2000.1173] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Doxorubicin (Dox), an anthracyclin antineoplastic agent, causes dilated cardiomyopathy. CARP has been identified as a nuclear protein whose mRNA levels are exquisitely sensitive to Dox. In this study we investigated the molecular mechanisms underlying the repression of CARP expression by Dox in cultured neonatal rat cardiac myocytes. Dox (1 micromol/l)-mediated decrease in CARP mRNA levels was strongly correlated with BNP but not with ANP mRNA levels. Hydrogen peroxide scavenger catalase (1 mg/ml) but not hydroxyl radical scavengers dimethylthiourea (10 mmol/l) or mannitol (10 mmol/l) blunted the Dox-mediated decrease in CARP and BNP expression. Superoxide dismutase inhibitor diethyldithiocarbamic acid (10 mmol/l), which inhibits the generation of hydrogen peroxide from superoxide metabolism, attenuated the repression. PD98059 (MEK1 inhibitor, 50 micromol/l), SB203580 (p38 MAP kinase inhibitor, 10 micromol/l), calphostin C (protein kinase C (PKC) inhibitor, 1 micromol/l), non-selective protein tyrosine kinase inhibitors genistein (50 micromol/l) or herbimycin A (1 micromol/l) failed to abrogate the downregulation of CARP and BNP expression by Dox. In contrast, H7 (30 micromol/l), a potent inhibitor of serine/threonine kinase, significantly blocked Dox-mediated downregulation of CARP and BNP expression. Transient transfection of a series of 5'-deletion and site-specific mutation constructs revealed that M-CAT element located at -37 of the human CARP promoter mediates Dox-induced repression of CARP promoter activity. These results suggest that a genetic response to Dox is mediated through the generation of hydrogen peroxide, which is selectively linked to the activation of H7-sensitive serine/threonine kinase distinct from PKC and well characterized mitogen-activated protein (MAP) kinases (ERK and p38MAP kinase). Furthermore, our data implicated M-CAT element as a Dox-response element within the CARP promoter in cardiac myocytes.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibiotics, Antineoplastic/pharmacology
- Antioxidants/pharmacology
- Base Sequence
- Cells, Cultured
- Dactinomycin/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation
- Doxorubicin/pharmacology
- Enzyme Inhibitors/pharmacology
- Humans
- Hydrogen Peroxide/metabolism
- Luciferases/metabolism
- MAP Kinase Signaling System
- Molecular Sequence Data
- Muscle Proteins
- Mutation
- Myocardium/metabolism
- Natriuretic Peptide, Brain/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nucleic Acid Synthesis Inhibitors/pharmacology
- Oxidative Stress/drug effects
- Plasmids/metabolism
- Promoter Regions, Genetic
- Protein Kinase C/metabolism
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Sequence Homology, Nucleic Acid
- Time Factors
- Transcription, Genetic/drug effects
- Transfection
Collapse
Affiliation(s)
- Y Aihara
- Second Department of Internal Medicine, Gunma University School of Medicine, Gunma, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Aihara Y, Kurabayashi M, Saito Y, Ohyama Y, Tanaka T, Takeda S, Tomaru K, Sekiguchi K, Arai M, Nakamura T, Nagai R. Cardiac ankyrin repeat protein is a novel marker of cardiac hypertrophy: role of M-CAT element within the promoter. Hypertension 2000; 36:48-53. [PMID: 10904011 DOI: 10.1161/01.hyp.36.1.48] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CARP, a cardiac doxorubicin (adriamycin)-responsive protein, has been identified as a nuclear protein whose expression is downregulated in response to doxorubicin. In the present study, we tested the hypothesis that CARP serves as a reliable genetic marker of cardiac hypertrophy in vivo and in vitro. CARP expression was markedly increased in 3 distinct models of cardiac hypertrophy in rats: constriction of abdominal aorta, spontaneously hypertensive rats, and Dahl salt-sensitive rats. In addition, we found that CARP mRNA levels correlate very strongly with the brain natriuretic peptide mRNA levels in Dahl rats. Transient transfection assays into primary cultures of neonatal rat cardiac myocytes indicate that transcription from the CARP and brain natriuretic peptide promoters is stimulated by overexpression of p38 and Rac1, components of the stress-activated mitogen-activated protein kinase pathways. Mutation analysis and electrophoretic mobility shift assays indicated that the M-CAT element can serve as a binding site for nuclear factors, and this element is important for the induction of CARP promoter activity by p38 and Rac1. Thus, our data suggest that M-CAT element is responsible for the regulation of the CARP gene in response to the activation of stress-responsive mitogen-activated protein kinase pathways. Moreover, given that activation of these pathways is associated with cardiac hypertrophy, we propose that CARP represents a novel genetic marker of cardiac hypertrophy.
Collapse
Affiliation(s)
- Y Aihara
- Second Department of Internal Medicine, Gunma University School of Medicine, Maebashi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ueyama T, Zhu C, Valenzuela YM, Suzow JG, Stewart AF. Identification of the functional domain in the transcription factor RTEF-1 that mediates alpha 1-adrenergic signaling in hypertrophied cardiac myocytes. J Biol Chem 2000; 275:17476-80. [PMID: 10764782 DOI: 10.1074/jbc.m001970200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiac myocytes respond to alpha(1)-adrenergic receptor stimulation by a progressive hypertrophy accompanied by the activation of many fetal genes, including skeletal muscle alpha-actin. The skeletal muscle alpha-actin gene is activated by signaling through an MCAT element, the binding site of the transcription enhancer factor-1 (TEF-1) family of transcription factors. Previously, we showed that overexpression of the TEF-1-related factor (RTEF-1) increased the alpha(1)-adrenergic response of the skeletal muscle alpha-actin promoter, whereas TEF-1 overexpression did not. Here, we identified the functional domains and specific sequences in RTEF-1 that mediate the alpha(1)-adrenergic response. Chimeric TEF-1 and RTEF-1 expression constructs localized the region responsible for the alpha(1)-adrenergic response to the carboxyl-terminal domain of RTEF-1. Site-directed mutagenesis was used to inactivate eight serine residues of RTEF-1, not present in TEF-1, that are putative targets of alpha(1)-adrenergic-dependent kinases. Mutation of a single serine residue, Ser-322, reduced the alpha(1)-adrenergic activation of RTEF-1 by 70% without affecting protein stability, suggesting that phosphorylation at this serine residue accounts for most of the alpha(1)-adrenergic response. Thus, these results demonstrate that RTEF-1 is a direct target of alpha(1)-adrenergic signaling in hypertrophied cardiac myocytes.
Collapse
Affiliation(s)
- T Ueyama
- Cardiovascular Institute, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | |
Collapse
|
33
|
He Q, Wu G, Lapointe MC. Isoproterenol and cAMP regulation of the human brain natriuretic peptide gene involves Src and Rac. Am J Physiol Endocrinol Metab 2000; 278:E1115-23. [PMID: 10827015 DOI: 10.1152/ajpendo.2000.278.6.e1115] [Citation(s) in RCA: 28] [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: 11/22/2022]
Abstract
Brain natriuretic peptide (BNP) gene expression and chronic activation of the sympathetic nervous system are characteristics of the development of heart failure. We studied the role of the beta-adrenergic signaling pathway in regulation of the human BNP (hBNP) promoter. An hBNP promoter (-1818 to +100) coupled to a luciferase reporter gene was transferred into neonatal cardiac myocytes, and luciferase activity was measured as an index of promoter activity. Isoproterenol (ISO), forskolin, and cAMP stimulated the promoter, and the beta(2)-antagonist ICI 118,551 abrogated the effect of ISO. In contrast, the protein kinase A (PKA) inhibitor H-89 failed to block the action of cAMP and ISO. Pertussis toxin (PT), which inactivates Galpha(i), inhibited ISO- and cAMP-stimulated hBNP promoter activity. The Src tyrosine kinase inhibitor PP1 and a dominant-negative mutant of the small G protein Rac also abolished the effect of ISO and cAMP. Finally, we studied the involvement of M-CAT-like binding sites in basal and inducible regulation of the hBNP promoter. Mutation of these elements decreased basal and cAMP-induced activity. These data suggest that beta-adrenergic regulation of hBNP is PKA independent, involves a Galpha(i)-activated pathway, and targets regulatory elements in the proximal BNP promoter.
Collapse
Affiliation(s)
- Q He
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
34
|
Calderone A, Abdelaziz N, Colombo F, Schreiber KL, Rindt H. A farnesyltransferase inhibitor attenuates cardiac myocyte hypertrophy and gene expression. J Mol Cell Cardiol 2000; 32:1127-40. [PMID: 10888263 DOI: 10.1006/jmcc.2000.1150] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The overexpression of either oncogenic ras or calmodulin in cardiac myocytes can elicit a hypertrophic response, albeit their recruitment by physiologically relevant stimuli remains unresolved. The present study utilized a pharmacological approach to examine the role of ras and calmodulin in norepinephrine- and endothelin-1-stimulated hypertrophy of neonatal rat cardiac myocytes. The pretreatment of cardiac myocytes with the farnesyltransferase inhibitor BMS-191563 (25 microM) increased the level of unfarnesylated ras in the cytosolic fraction, and caused a concomitant 42 +/- 2% decrease in immunodetectable farnesylated ras in the particulate fraction. In parallel, BMS-191563 pretreatment inhibited norepinephrine-mediated 3H-leucine uptake (80 +/- 10% decrease: n = 6; P<0.01), whereas a significant but less pronounced effect on the endothelin-1 response (46 +/- 6% decrease: n = 6; P<0.05) was observed. The calmodulin inhibitor W7 caused a 50 +/- 10% decrease (n = 8; P<0.05) of norepinephrine stimulated protein synthesis, whereas the endothelin-1 response was unaffected. Consistent with the recruitment of ras, BMS-191563 pretreatment attenuated norepinephrine and endothelin-1-stimulated extracellular signal-regulated kinase (ERK) activity. However, PD098059-mediated inhibition of MEK-dependent stimulation of ERK did not alter the hypertrophic response of either agonist. At the molecular level, the pretreatment with either BMS-191563 or W7 attenuated the norepinephrine-mediated increase of prepro-ANP and -BNP mRNA. Likewise, BMS-191563 caused a significant decrease of endothelin-1-mediated expression of the natriuretic peptide mRNAs, but to a lesser extent, as compared to norepinephrine. Thus, the present study has shown the treatment of neonatal rat cardiac myocytes with a farnesyltransferase inhibitor can attenuate the hypertrophic phenotype in response to physiologically relevant stimuli, thereby supporting a role of the small GTP-binding protein ras. Moreover, these data further suggest alternative ras-independent signaling pathways are also implicated in the hypertrophic response, albeit, there appears to exist a stimulus-specific heterogeneity in their recruitment.
Collapse
Affiliation(s)
- A Calderone
- Université de Montréal, Departément de Physiologie et Biochimie, et l'Institut de Cardiologie de Montréal, Québec, Canada.
| | | | | | | | | |
Collapse
|
35
|
Takei Y. Structural and functional evolution of the natriuretic peptide system in vertebrates. INTERNATIONAL REVIEW OF CYTOLOGY 1999; 194:1-66. [PMID: 10494624 DOI: 10.1016/s0074-7696(08)62394-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The natriuretic peptide (NP) system consists of three types of hormones [atrial NP (ANP), brain or B-type NP (BNP), and C-type NP (CNP)] and three types of receptors [NP receptor (R)-A, NPR-B, and NPR-C]. ANP and BNP are circulating hormones secreted from the heart, whereas CNP is basically a neuropeptide. NPR-A and NPR-B are membrane-bound guanylyl cyclases, whereas NPR-C is assumed to function as a clearance-type receptor. ANP, BNP, and CNP occur commonly in all tetrapods, but ventricular NP replaces BNP in teleost fish. In elasmobranchs, only CNP is found, even in the heart, suggesting that CNP is an ancestral form. A new guanylyl cyclase-uncoupled receptor named NPR-D has been identified in the eel in addition to NPR-A, -B, and -C. The NP system plays pivotal roles in cardiovascular and body fluid homeostasis. ANP is secreted in response to an increase in blood volume and acts on various organs to decrease both water and Na+, resulting in restoration of blood volume. In the eel, however, ANP is secreted in response to an increase in plasma osmolality and decreases Na+ specifically, thereby promoting seawater adaptation. Therefore, it seems that the family of NPs were originally Na(+)-extruding hormones in fishes; however, they evolved to be volume-depleting hormones promoting the excretion of both Na+ and water in tetrapods in which both are always regulated in the same direction. Vertebrates expanded their habitats from fresh water to the sea or to land during evolution. The structure and function of osmoregulatory hormones have also undergone evolution during this ecological evolution. Thus, a comparative approach to the study of the NP family affords new insights into the essential function of this osmoregulatory hormone.
Collapse
Affiliation(s)
- Y Takei
- Laboratory of Physiology, University of Tokyo, Japan
| |
Collapse
|
36
|
Morimoto T, Hasegawa K, Kaburagi S, Kakita T, Masutani H, Kitsis RN, Matsumori A, Sasayama S. GATA-5 is involved in leukemia inhibitory factor-responsive transcription of the beta-myosin heavy chain gene in cardiac myocytes. J Biol Chem 1999; 274:12811-8. [PMID: 10212267 DOI: 10.1074/jbc.274.18.12811] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukemia inhibitory factor is a member of a family of structurally related cytokines sharing the receptor component gp130. Activation of gp130 by leukemia inhibitory factor is sufficient to induce myocardial cell hypertrophy accompanied by specific changes in the pattern of gene expression. However, the molecular mechanisms that link gp130 activation to these changes have not been clarified. The present study investigated the transcriptional pathways by which leukemia inhibitory factor activates beta-myosin heavy chain expression during myocardial cell hypertrophy. Mutation of the GATA motif in the beta-myosin heavy chain promoter totally abolished leukemia inhibitory factor-responsive transcription without changing basal transcriptional activity. In contrast, endothelin-1 responsiveness was unaffected by the GATA mutation. Among members of the cardiac GATA transcription factor subfamily (GATA-4, -5, and -6), GATA-5 was the sole and potent transactivator for the beta-myosin heavy chain promoter. This transactivation was dependent on sequence-specific binding of GATA-5 to the beta-myosin heavy chain GATA element. Cardiac nuclear factors that bind to to the beta-myosin heavy chain GATA element were induced by leukemia inhibitory factor stimulation. Last, leukemia inhibitory factor stimulation markedly increased transcripts of cardiac GATA-5, the expression of which is normally restricted to the early embryo. Thus, GATA-5 may be involved in gp130 signaling in cardiac myocytes.
Collapse
Affiliation(s)
- T Morimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
He Q, LaPointe MC. Interleukin-1beta regulation of the human brain natriuretic peptide promoter involves Ras-, Rac-, and p38 kinase-dependent pathways in cardiac myocytes. Hypertension 1999; 33:283-9. [PMID: 9931118 DOI: 10.1161/01.hyp.33.1.283] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Because both the brain natriuretic peptide (BNP) gene and the cytokine interleukin-1beta (IL-1beta) are induced in the infarcted myocardium, localized production of IL-1beta may regulate the BNP gene. We tested whether (1) IL-1beta regulates the human BNP promoter, (2) cis elements in the proximal promoter respond to IL-1beta, and (3) mitogen-activated protein kinase (MAPK) signaling pathways [p42/44, c-jun (JNK) and p38 kinase] are involved. We transferred the hBNP promoter coupled to a luciferase reporter gene or constructs with mutations in the proximal promoter GATA and M-CAT elements into neonatal rat ventricular myocytes and treated the cells with IL-1beta for 24 hours. IL-1beta-stimulated hBNP luciferase activity was eliminated by pretreatment with the transcription inhibitor actinomycin D. Both the p38 kinase inhibitor SB205380 (SB) and cotransfection of a dominant-negative mutant of p38 kinase reduced IL-1beta stimulation of the hBNP promoter. Dominant-negative mutants of Ras and Rac inhibited IL-1beta-stimulated hBNP luciferase activity by 64% and 90%, respectively. Constitutively active forms of Rac and MKK6, the immediate upstream activator of p38, were stimulatory; however, only the effect of MKK6 was inhibited by SB. Neither the p42/44 nor the JNK pathway was involved in the action of IL-1beta. Both IL-1beta and MKK6 activation of the hBNP promoter were partially reduced when the promoter contained a mutated M-CAT element. In summary, (1) IL-1beta is a transcriptional activator of the hBNP promoter; (2) IL-1beta acts through a Ras-dependent pathway not coupled to activation of p42/44 MAPK or JNK; (3) IL-1beta acts through a Rac-dependent pathway, but the downstream effector is not known; and (4) IL-1beta activation of p38 kinase is partially involved in regulation of the hBNP promoter, targeting the proximal M-CAT element.
Collapse
Affiliation(s)
- Q He
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, Mich.48202-2689, USA
| | | |
Collapse
|
38
|
Kovacic B, Ilić D, Damsky CH, Gardner DG. c-Src activation plays a role in endothelin-dependent hypertrophy of the cardiac myocyte. J Biol Chem 1998; 273:35185-93. [PMID: 9857056 DOI: 10.1074/jbc.273.52.35185] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of the atrial natriuretic peptide (ANP) gene is regarded as one of the earliest and most reliable markers of hypertrophy in the ventricular cardiac myocyte. We have examined the role of the nonreceptor tyrosine kinases in the signaling mechanism(s) leading to hypertrophy using human ANP gene promoter activity as a marker. Endothelin (ET), a well known hypertrophic agonist, increased activity of c-Src, c-Yes, and Fyn within minutes and promoted a selective redistribution of each of these kinases within the cell. Overexpression of c-Src effected a significant increase in activity of a cotransfected human ANP promoter-driven chloramphenicol acetyl transferase reporter, while expression of either c-Yes or Fyn was considerably less effective in this regard. ET-dependent stimulation of the human ANP gene promoter was partially inhibited by co-transfection with dominant negative Ras or dominant negative Src or Csk or by treatment with the potent Src family-selective tyrosine kinase inhibitor PP1, suggesting that the Src family kinases are involved in signaling ET-dependent activation of this promoter. Both ET- and Src-dependent activation of the ANP promoter required the presence of a CArG motif in a serum response element-like structure between -422 and -413 but did not appear to require assembly of a ternary complex for full activity. These findings support a role for Src in the activation of ANP gene expression and suggest that this kinase may contribute in an important way to the signaling mechanisms that activate hypertrophy in the cardiac myocyte.
Collapse
Affiliation(s)
- B Kovacic
- Metabolic Research Unit and Departments of Medicine, University of California, San Francisco, California 94143, USA.
| | | | | | | |
Collapse
|
39
|
Thuerauf DJ, Arnold ND, Zechner D, Hanford DS, DeMartin KM, McDonough PM, Prywes R, Glembotski CC. p38 Mitogen-activated protein kinase mediates the transcriptional induction of the atrial natriuretic factor gene through a serum response element. A potential role for the transcription factor ATF6. J Biol Chem 1998; 273:20636-43. [PMID: 9685422 DOI: 10.1074/jbc.273.32.20636] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In various cell types certain stresses can stimulate p38 mitogen-activated protein kinase (p38 MAPK), leading to the transcriptional activation of genes that contribute to appropriate compensatory responses. In this report the mechanism of p38-activated transcription was studied in cardiac myocytes where this MAPK is a key regulator of the cell growth and the cardiac-specific gene induction that occurs in response to potentially stressful stimuli. In the cardiac atrial natriuretic factor (ANF) gene, a promoter-proximal serum response element (SRE), which binds serum response factor (SRF), was shown to be critical for ANF induction in primary cardiac myocytes transfected with the selective p38 MAPK activator, MKK6 (Glu). This ANF SRE does not possess sequences typically required for the binding of the Ets-related ternary complex factors (TCFs), such as Elk-1, indicating that p38-mediated induction through this element may take place independently of such TCFs. Although p38 did not phosphorylate SRF in vitro, it efficiently phosphorylated ATF6, a newly discovered SRF-binding protein that is believed to serve as a co-activator of SRF-inducible transcription at SREs. Expression of an ATF6 antisense RNA blocked p38-mediated ANF induction through the ANF SRE. Moreover, when fused to the Gal4 DNA-binding domain, an N-terminal 273-amino acid fragment of ATF6 was sufficient to support trans-activation of Gal4/luciferase expression in response to p38 but not the other stress kinase, N-terminal Jun kinase (JNK); p38-activating cardiac growth promoters also stimulated ATF6 trans-activation. These results indicate that through ATF6, p38 can augment SRE-mediated transcription independently of Ets-related TCFs, representing a novel mechanism of SRF-dependent transcription by MAP kinases.
Collapse
Affiliation(s)
- D J Thuerauf
- Department of Biology and Molecular Biology, Institute, San Diego State University, San Diego, California 92182, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Stewart AF, Suzow J, Kubota T, Ueyama T, Chen HH. Transcription factor RTEF-1 mediates alpha1-adrenergic reactivation of the fetal gene program in cardiac myocytes. Circ Res 1998; 83:43-9. [PMID: 9670917 DOI: 10.1161/01.res.83.1.43] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alpha1-adrenergic receptor stimulation induces cardiac myocytes to hypertrophy and reactivates many fetal genes, including beta-myosin heavy chain (betaMyHC) and skeletal alpha-actin (SKA), by signaling through myocyte-specific CAT (M-CAT) cis elements, binding sites of the transcriptional enhancer factor-1 (TEF-1) family of transcription factors. To examine functional differences between TEF-1 and related to TEF-1 (RTEF-1) in alpha1-adrenergic reactivation of the fetal program, expression constructs were cotransfected with betaMyHC and SKA promoter/reporter constructs in neonatal rat cardiac myocytes. TEF-I overexpression tended to transactivate a minimal betaMyHC promoter but significantly interfered with a minimal SKA promoter. In contrast, RTEF-1 transactivated both the minimal betaMyHC and SKA promoters. TEF-1 and RTEF-I also affected the alpha1-adrenergic response of the betaMyHC and SKA promoters differently. TEF-1 had no effect. In contrast, RTEF-1 potentiated the alpha1-adrenergic responses of the SKA promoter and of a -3.3-kb betaMyHC promoter. To determine why the promoters responded differently to TEF-1 and RTEF-1, promoters with mutated M-CAT elements were tested in the same way. The betaMyHC promoter required an intact M-CAT element to respond to TEF-1 and RTEF-1, whereas the SKA promoter M-CAT was required for the TEF-1 response but not for the RTEF-1 response, suggesting that SKA promoter-specific cofactors may be involved. By competition gel shift assay, the M-CAT of the minimal betaMyHC promoter had a lower affinity than that of the SKA promoter, which partly explains the different responses of these promoters to TEF-1. These results highlight functional differences between TEF-1 and RTEF-1 and suggest a novel function of RTEF-1 in mediating the alpha1-adrenergic response in hypertrophic cardiac myocytes.
Collapse
Affiliation(s)
- A F Stewart
- Department of Medicine, University of Pittsburgh, PA 15213, USA. als6+@pitt.edu
| | | | | | | | | |
Collapse
|
41
|
Zechner D, Craig R, Hanford DS, McDonough PM, Sabbadini RA, Glembotski CC. MKK6 activates myocardial cell NF-kappaB and inhibits apoptosis in a p38 mitogen-activated protein kinase-dependent manner. J Biol Chem 1998; 273:8232-9. [PMID: 9525929 DOI: 10.1074/jbc.273.14.8232] [Citation(s) in RCA: 177] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In cardiac myocytes the stimulation of p38 mitogen-activated protein kinase activates a hypertrophic growth program and the induction of the cardiac-specific genes associated with this program. This study focused on determining whether these novel growth-promoting effects are accompanied by the p38-mediated inhibition of apoptosis, and if so, what signaling pathways might be responsible. Primary neonatal rat ventricular myocytes were driven into apoptosis by treatments known to induce apoptosis in other cell types, e.g. incubation with anisomycin or overexpression constitutively active MEKK-1 (MEKK-1COOH), a protein that strongly activates extracellular signal-regulated kinase and N-terminal c-Jun kinase, but not p38. Overexpression of constitutively active MKK6, MKK6 (Glu), which selectively activates p38 in cardiac myocytes, protected cells from either anisomycin- or MEKK-1COOH-induced apoptosis. This protection was blocked by SB 203580, a selective p38 inhibitor. MKK6 (Glu) also activated transcription mediated by NF-kappaB, a factor which protects other cell types from apoptosis. The activation of NF-kappaB and the protection from apoptosis mediated by MKK6 (Glu) were both blocked by SB 203580. Interestingly, overexpression of a mutant form of I-kappaBalpha, which inhibits nuclear translocation of NF-kappaB, completely blocked MKK6 (Glu)-activated NF-kappaB but had little effect on MKK6s anti-apoptotic effects. These findings suggest that, in part, the overexpression of MKK6 (Glu) may foster growth and survival of cardiac myocytes by protecting them from apoptosis in a p38-dependent manner. Additionally, while NF-kappaB is activated in myocardial cells by p38, this does not appear to be the major mechanism by which MKK6 (Glu) exerts its anti-apoptotic effects in this cell type, suggesting a novel pathway for p38-mediated protection from apoptosis.
Collapse
Affiliation(s)
- D Zechner
- Department of Biology and Molecular Biology Institute, San Diego State University, San Diego, California 92182, USA
| | | | | | | | | | | |
Collapse
|
42
|
Zechner D, Thuerauf DJ, Hanford DS, McDonough PM, Glembotski CC. A role for the p38 mitogen-activated protein kinase pathway in myocardial cell growth, sarcomeric organization, and cardiac-specific gene expression. J Cell Biol 1997; 139:115-27. [PMID: 9314533 PMCID: PMC2139826 DOI: 10.1083/jcb.139.1.115] [Citation(s) in RCA: 251] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/1997] [Revised: 07/09/1997] [Indexed: 02/05/2023] Open
Abstract
Three hallmark features of the cardiac hypertrophic growth program are increases in cell size, sarcomeric organization, and the induction of certain cardiac-specific genes. All three features of hypertrophy are induced in cultured myocardial cells by alpha1- adrenergic receptor agonists, such as phenylephrine (PE) and other growth factors that activate mitogen- activated protein kinases (MAPKs). In this study the MAPK family members extracellular signal-regulated kinase (ERK), c-jun NH2-terminal kinase (JNK), and p38 were activated by transfecting cultured cardiac myocytes with constructs encoding the appropriate kinases possessing gain-of-function mutations. Transfected cells were then analyzed for changes in cell size, sarcomeric organization, and induction of the genes for the A- and B-type natriuretic peptides (NPs), as well as the alpha-skeletal actin (alpha-SkA) gene. While activation of JNK and/or ERK with MEKK1COOH or Raf-1 BXB, respectively, augmented cell size and effected relatively modest increases in NP and alpha-SkA promoter activities, neither upstream kinase conferred sarcomeric organization. However, transfection with MKK6 (Glu), which specifically activated p38, augmented cell size, induced NP and alpha-Ska promoter activities by up to 130-fold, and elicited sarcomeric organization in a manner similar to PE. Moreover, all three growth features induced by MKK6 (Glu) or PE were blocked with the p38-specific inhibitor, SB 203580. These results demonstrate novel and potentially central roles for MKK6 and p38 in the regulation of myocardial cell hypertrophy.
Collapse
Affiliation(s)
- D Zechner
- Department of Biology and Molecular Biology Institute, San Diego State University, California 92182, USA
| | | | | | | | | |
Collapse
|