1
|
Triposkiadis F, Briasoulis A, Sarafidis P, Magouliotis D, Athanasiou T, Paraskevaidis I, Skoularigis J, Xanthopoulos A. The Sympathetic Nervous System in Hypertensive Heart Failure with Preserved LVEF. J Clin Med 2023; 12:6486. [PMID: 37892623 PMCID: PMC10607346 DOI: 10.3390/jcm12206486] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The neurohormonal model of heart failure (HF) pathogenesis states that a reduction in cardiac output caused by cardiac injury results in sympathetic nervous system (SNS) activation, that is adaptive in the short-term and maladaptive in the long-term. This model has proved extremely valid and has been applied in HF with a reduced left ventricular (LV) ejection fraction (LVEF). In contrast, it has been undermined in HF with preserved LVEF (HFpEF), which is due to hypertension (HTN) in the vast majority of the cases. Erroneously, HTN, which is the leading cause of cardiovascular disease and premature death worldwide and is present in more than 90% of HF patients, is tightly linked with SNS overactivity. In this paper we provide a contemporary overview of the contribution of SNS overactivity to the development and progression of hypertensive HF (HHF) as well as the clinical implications resulting from therapeutic interventions modifying SNS activity. Throughout the manuscript the terms HHF with preserved LVEF and HfpEF will be used interchangeably, considering that the findings in most HFpEF studies are driven by HTN.
Collapse
Affiliation(s)
| | - Alexandros Briasoulis
- Department of Therapeutics, Heart Failure and Cardio-Oncology Clinic, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Dimitrios Magouliotis
- Unit of Quality Improvement, Department of Cardiothoracic Surgery, University of Thessaly, 411 10 Biopolis, Greece;
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary’s Hospital, London W2 1NY, UK;
| | | | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, 411 10 Larissa, Greece;
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 411 10 Larissa, Greece;
| |
Collapse
|
2
|
Mechanotransduction of mesenchymal stem cells (MSCs) during cardiomyocytes differentiation. Heliyon 2022; 8:e11624. [DOI: 10.1016/j.heliyon.2022.e11624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/15/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
|
3
|
Hynynen H, Mutikainen M, Naumenko N, Shakirzyanova A, Tuomainen T, Tavi P. Short high-fat diet interferes with the physiological maturation of the late adolescent mouse heart. Physiol Rep 2020; 8:e14474. [PMID: 32643294 PMCID: PMC7343666 DOI: 10.14814/phy2.14474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/12/2020] [Accepted: 05/04/2020] [Indexed: 11/24/2022] Open
Abstract
Dietary fats are essential for cardiac function. The metabolites of fats known as fatty acids provide most of the energy for cardiac tissue, serve as building blocks for membranes and regulate important signaling cascades. Despite their importance, excess fat intake can cause cardiac dysfunction. The detrimental effects of high-fat diet (HFD) on cardiac health are widely investigated in long-term studies but the short-term effects of fats have not been thoroughly studied. To elucidate the near-term effects of a HFD on the growth and maturation of late adolescent heart we subjected 11-week-old mice to an 8-week long HFD (42% of calories from fat, 42% from carbohydrate, n = 8) or chow diet (12% of calories from fat, 66% from carbohydrate, n = 7) and assessed their effects on the heart in vivo and in vitro. Our results showed that excessive fat feeding interferes with normal maturation of the heart indicated by the lack of increase in dimensions, volume, and stroke volume of the left ventricles of mice on high fat diet that were evident in mice on chow diet. In addition, differences in regional strain during the contraction cycle between mice on HFD and chow diet were seen. These changes were associated with reduced activity of the growth promoting PI3K-Akt1 signaling cascade and moderate changes in glucose metabolism without changes in calcium signaling. This study suggests that even a short period of HFD during late adolescence hinders cardiac maturation and causes physiological changes that may have an impact on the cardiac health in adulthood.
Collapse
Affiliation(s)
- Heidi Hynynen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Maija Mutikainen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Nikolay Naumenko
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | | | - Tomi Tuomainen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Pasi Tavi
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| |
Collapse
|
4
|
Gaetani R, Zizzi EA, Deriu MA, Morbiducci U, Pesce M, Messina E. When Stiffness Matters: Mechanosensing in Heart Development and Disease. Front Cell Dev Biol 2020; 8:334. [PMID: 32671058 PMCID: PMC7326078 DOI: 10.3389/fcell.2020.00334] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
During embryonic morphogenesis, the heart undergoes a complex series of cellular phenotypic maturations (e.g., transition of myocytes from proliferative to quiescent or maturation of the contractile apparatus), and this involves stiffening of the extracellular matrix (ECM) acting in concert with morphogenetic signals. The maladaptive remodeling of the myocardium, one of the processes involved in determination of heart failure, also involves mechanical cues, with a progressive stiffening of the tissue that produces cellular mechanical damage, inflammation, and ultimately myocardial fibrosis. The assessment of the biomechanical dependence of the molecular machinery (in myocardial and non-myocardial cells) is therefore essential to contextualize the maturation of the cardiac tissue at early stages and understand its pathologic evolution in aging. Because systems to perform multiscale modeling of cellular and tissue mechanics have been developed, it appears particularly novel to design integrated mechano-molecular models of heart development and disease to be tested in ex vivo reconstituted cells/tissue-mimicking conditions. In the present contribution, we will discuss the latest implication of mechanosensing in heart development and pathology, describe the most recent models of cell/tissue mechanics, and delineate novel strategies to target the consequences of heart failure with personalized approaches based on tissue engineering and induced pluripotent stem cell (iPSC) technologies.
Collapse
Affiliation(s)
- Roberto Gaetani
- Department of Molecular Medicine, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Rome, Italy.,Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, San Diego, CA, United States
| | - Eric Adriano Zizzi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Marco Agostino Deriu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Umberto Morbiducci
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino," IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Maternal, Infantile, and Urological Sciences, "Umberto I" Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Krueger W, Bender N, Haeusler M, Henneberg M. The role of mechanotransduction in heart failure pathobiology-a concise review. Heart Fail Rev 2020; 26:981-995. [PMID: 31965473 DOI: 10.1007/s10741-020-09915-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This review evaluates the role of mechanotransduction (MT) in heart failure (HF) pathobiology. Cardiac functional and structural modifications are regulated by biomechanical forces. Exposing cardiomyocytes and the myocardial tissue to altered biomechanical stress precipitates changes in the end-diastolic wall stress (EDWS). Thereby various interconnected biomolecular pathways, essentially mediated and orchestrated by MT, are launched and jointly contribute to adapt and remodel the myocardium. This cardiac MT-mediated feedback decisively determines the primary cardiac cellular and tissue response, the sort (concentric or eccentric) of hypertrophy/remodeling, to mechanical and/or hemodynamic alterations. Moreover, the altered EDWS affects the diastolic myocardial properties independent of the systolic function, and elevated EDWS causes diastolic dysfunction. The close interconnection between MT pathways and the cell nucleus, the genetic endowment, principally allows for the wide variety of phenotypic appearances. However, demographic, environmental features, comorbidities, and also the genetic make-up may modulate the phenotypic result. Cardiac MT takes a fundamental and superordinate position in the myocardial adaptation and remodeling processes in all HF categories and phenotypes. Therefore, the effects of MT should be integrated in all our scientific, clinical, and therapeutic considerations.
Collapse
Affiliation(s)
- Wolfgang Krueger
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland. .,Medical University Department, Kantonsspital Aarau, Aarau, Switzerland.
| | - Nicole Bender
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Martin Haeusler
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Maciej Henneberg
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
6
|
ERK: A Key Player in the Pathophysiology of Cardiac Hypertrophy. Int J Mol Sci 2019; 20:ijms20092164. [PMID: 31052420 PMCID: PMC6539093 DOI: 10.3390/ijms20092164] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiac hypertrophy is an adaptive and compensatory mechanism preserving cardiac output during detrimental stimuli. Nevertheless, long-term stimuli incite chronic hypertrophy and may lead to heart failure. In this review, we analyze the recent literature regarding the role of ERK (extracellular signal-regulated kinase) activity in cardiac hypertrophy. ERK signaling produces beneficial effects during the early phase of chronic pressure overload in response to G protein-coupled receptors (GPCRs) and integrin stimulation. These functions comprise (i) adaptive concentric hypertrophy and (ii) cell death prevention. On the other hand, ERK participates in maladaptive hypertrophy during hypertension and chemotherapy-mediated cardiac side effects. Specific ERK-associated scaffold proteins are implicated in either cardioprotective or detrimental hypertrophic functions. Interestingly, ERK phosphorylated at threonine 188 and activated ERK5 (the big MAPK 1) are associated with pathological forms of hypertrophy. Finally, we examine the connection between ERK activation and hypertrophy in (i) transgenic mice overexpressing constitutively activated RTKs (receptor tyrosine kinases), (ii) animal models with mutated sarcomeric proteins characteristic of inherited hypertrophic cardiomyopathies (HCMs), and (iii) mice reproducing syndromic genetic RASopathies. Overall, the scientific literature suggests that during cardiac hypertrophy, ERK could be a “good” player to be stimulated or a “bad” actor to be mitigated, depending on the pathophysiological context.
Collapse
|
7
|
Johnson DM, Antoons G. Arrhythmogenic Mechanisms in Heart Failure: Linking β-Adrenergic Stimulation, Stretch, and Calcium. Front Physiol 2018; 9:1453. [PMID: 30374311 PMCID: PMC6196916 DOI: 10.3389/fphys.2018.01453] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Heart failure (HF) is associated with elevated sympathetic tone and mechanical load. Both systems activate signaling transduction pathways that increase cardiac output, but eventually become part of the disease process itself leading to further worsening of cardiac function. These alterations can adversely contribute to electrical instability, at least in part due to the modulation of Ca2+ handling at the level of the single cardiac myocyte. The major aim of this review is to provide a definitive overview of the links and cross talk between β-adrenergic stimulation, mechanical load, and arrhythmogenesis in the setting of HF. We will initially review the role of Ca2+ in the induction of both early and delayed afterdepolarizations, the role that β-adrenergic stimulation plays in the initiation of these and how the propensity for these may be altered in HF. We will then go onto reviewing the current data with regards to the link between mechanical load and afterdepolarizations, the associated mechano-sensitivity of the ryanodine receptor and other stretch activated channels that may be associated with HF-associated arrhythmias. Furthermore, we will discuss how alterations in local Ca2+ microdomains during the remodeling process associated the HF may contribute to the increased disposition for β-adrenergic or stretch induced arrhythmogenic triggers. Finally, the potential mechanisms linking β-adrenergic stimulation and mechanical stretch will be clarified, with the aim of finding common modalities of arrhythmogenesis that could be targeted by novel therapeutic agents in the setting of HF.
Collapse
Affiliation(s)
- Daniel M Johnson
- Department of Cardiothoracic Surgery, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Gudrun Antoons
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
8
|
Kennedy-Lydon T, Rosenthal N. Cardiac regeneration: All work and no repair? Sci Transl Med 2017; 9:9/383/eaad9019. [PMID: 28356512 DOI: 10.1126/scitranslmed.aad9019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 12/19/2016] [Indexed: 12/13/2022]
Abstract
Structural changes in the developing heart may influence the limited regenerative capacity of the adult heart. We examine how the workload exerted on the adult mammalian heart may limit regenerative capability and discuss recent therapies that demonstrate beneficial effects through unloading the heart.
Collapse
Affiliation(s)
| | - Nadia Rosenthal
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,The Jackson Laboratory, Bar Harbor, ME, USA
| |
Collapse
|
9
|
Lottonen-Raikaslehto L, Rissanen R, Gurzeler E, Merentie M, Huusko J, Schneider JE, Liimatainen T, Ylä-Herttuala S. Left ventricular remodeling leads to heart failure in mice with cardiac-specific overexpression of VEGF-B 167: echocardiography and magnetic resonance imaging study. Physiol Rep 2017; 5:5/6/e13096. [PMID: 28351964 PMCID: PMC5371547 DOI: 10.14814/phy2.13096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 10/07/2016] [Accepted: 11/20/2016] [Indexed: 01/24/2023] Open
Abstract
Cardiac-specific overexpression of vascular endothelial growth factor (VEGF)-B167 is known to induce left ventricular hypertrophy due to altered lipid metabolism, in which ceramides accumulate to the heart and cause mitochondrial damage. The aim of this study was to evaluate and compare different imaging methods to find the most sensitive way to diagnose at early stage the progressive left ventricular remodeling leading to heart failure. Echocardiography and cardiovascular magnetic resonance imaging were compared for imaging the hearts of transgenic mice with cardiac-specific overexpression of VEGF-B167 and wild-type mice from 5 to 14 months of age at several time points. Disease progression was verified by molecular biology methods and histology. We showed that left ventricular remodeling is already ongoing at the age of 5 months in transgenic mice leading to heart failure by the age of 14 months. Measurements from echocardiography and cardiovascular magnetic resonance imaging revealed similar changes in cardiac structure and function in the transgenic mice. Changes in histology, gene expressions, and electrocardiography supported the progression of left ventricular hypertrophy. Longitudinal relaxation time in rotating frame (T1ρ ) in cardiovascular magnetic resonance imaging could be suitable for detecting severe fibrosis in the heart. We conclude that cardiac-specific overexpression of VEGF-B167 leads to left ventricular remodeling at early age and is a suitable model to study heart failure development with different imaging methods.
Collapse
Affiliation(s)
- Line Lottonen-Raikaslehto
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riina Rissanen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Erika Gurzeler
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mari Merentie
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jenni Huusko
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jurgen E Schneider
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, United kingdom
| | - Timo Liimatainen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.,Clinical Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland .,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland.,Heart Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
10
|
Tuomainen T, Tavi P. The role of cardiac energy metabolism in cardiac hypertrophy and failure. Exp Cell Res 2017; 360:12-18. [DOI: 10.1016/j.yexcr.2017.03.052] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 12/22/2022]
|
11
|
Colak D, Alaiya AA, Kaya N, Muiya NP, AlHarazi O, Shinwari Z, Andres E, Dzimiri N. Integrated Left Ventricular Global Transcriptome and Proteome Profiling in Human End-Stage Dilated Cardiomyopathy. PLoS One 2016; 11:e0162669. [PMID: 27711126 PMCID: PMC5053516 DOI: 10.1371/journal.pone.0162669] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 08/28/2016] [Indexed: 01/30/2023] Open
Abstract
Aims The disease pathways leading to idiopathic dilated cardiomyopathy (DCM) are still elusive. The present study investigated integrated global transcriptional and translational changes in human DCM for disease biomarker discovery. Methods We used identical myocardial tissues from five DCM hearts compared to five non-failing (NF) donor hearts for both transcriptome profiling using the ABI high-density oligonucleotide microarrays and proteome expression with One-Dimensional Nano Acquity liquid chromatography coupled with tandem mass spectrometry on the Synapt G2 system. Results We identified 1262 differentially expressed genes (DEGs) and 269 proteins (DEPs) between DCM cases and healthy controls. Among the most significantly upregulated (>5-fold) proteins were GRK5, APOA2, IGHG3, ANXA6, HSP90AA1, and ATP5C1 (p< 0.01). On the other hand, the most significantly downregulated proteins were GSTM5, COX17, CAV1 and ANXA3. At least ten entities were concomitantly upregulated on the two analysis platforms: GOT1, ALDH4A1, PDHB, BDH1, SLC2A11, HSP90AA1, HSP90AB1, H2AFV, HSPA5 and NDUFV1. Gene ontology analyses of DEGs and DEPs revealed significant overlap with enrichment of genes/proteins related to metabolic process, biosynthetic process, cellular component organization, oxidative phosphorylation, alterations in glycolysis and ATP synthesis, Alzheimer’s disease, chemokine-mediated inflammation and cytokine signalling pathways. Conclusion The concomitant use of transcriptome and proteome expression to evaluate global changes in DCM has led to the identification of sixteen commonly altered entities as well as novel genes, proteins and pathways whose cardiac functions have yet to be deciphered. This data should contribute towards better management of the disease.
Collapse
Affiliation(s)
- Dilek Colak
- Biostatistics, Epidemiology and Scientific Computing Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Ayodele A. Alaiya
- Proteomics Unit, Stem Cell Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Namik Kaya
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Nzioka P. Muiya
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Olfat AlHarazi
- Biostatistics, Epidemiology and Scientific Computing Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Zakia Shinwari
- Proteomics Unit, Stem Cell Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Editha Andres
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Nduna Dzimiri
- Genetics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
- * E-mail:
| |
Collapse
|
12
|
Koivisto E, Jurado Acosta A, Moilanen AM, Tokola H, Aro J, Pennanen H, Säkkinen H, Kaikkonen L, Ruskoaho H, Rysä J. Characterization of the regulatory mechanisms of activating transcription factor 3 by hypertrophic stimuli in rat cardiomyocytes. PLoS One 2014; 9:e105168. [PMID: 25136830 PMCID: PMC4138181 DOI: 10.1371/journal.pone.0105168] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/18/2014] [Indexed: 01/08/2023] Open
Abstract
Aims Activating transcription factor 3 (ATF3) is a stress-activated immediate early gene suggested to have both detrimental and cardioprotective role in the heart. Here we studied the mechanisms of ATF3 activation by hypertrophic stimuli and ATF3 downstream targets in rat cardiomyocytes. Methods and Results When neonatal rat cardiomyocytes were exposed to endothelin-1 (ET-1, 100 nM) and mechanical stretching in vitro, maximal increase in ATF3 expression occurred at 1 hour. Inhibition of extracellular signal-regulated kinase (ERK) by PD98059 decreased ET-1– and stretch–induced increase of ATF3 protein but not ATF3 mRNA levels, whereas protein kinase A (PKA) inhibitor H89 attenuated both ATF3 mRNA transcription and protein expression in response to ET-1 and stretch. To characterize further the regulatory mechanisms upstream of ATF3, p38 mitogen-activated protein kinase (MAPK) signaling was investigated using a gain-of-function approach. Adenoviral overexpression of p38α, but not p38β, increased ATF3 mRNA and protein levels as well as DNA binding activity. To investigate the role of ATF3 in hypertrophic process, we overexpressed ATF3 by adenovirus-mediated gene transfer. In vitro, ATF3 gene delivery attenuated the mRNA transcription of interleukin-6 (IL-6) and plasminogen activator inhibitor-1 (PAI-1), and enhanced nuclear factor-κB (NF-κB) and Nkx-2.5 DNA binding activities. Reduced PAI-1 expression was also detected in vivo in adult rat heart by direct intramyocardial adenovirus-mediated ATF3 gene delivery. Conclusions These data demonstrate that ATF3 activation by ET-1 and mechanical stretch is partly mediated through ERK and cAMP-PKA pathways, whereas p38 MAPK pathway is involved in ATF3 activation exclusively through p38α isoform. ATF3 activation caused induction of modulators of the inflammatory response NF-κB and Nkx-2.5, as well as attenuation of pro-fibrotic and pro-inflammatory proteins IL-6 and PAI-1, suggesting cardioprotective role for ATF3 in the heart.
Collapse
Affiliation(s)
- Elina Koivisto
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Alicia Jurado Acosta
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Anne-Mari Moilanen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Department of Pathology, Institute of Diagnostics, University of Oulu, Oulu, Finland
| | - Heikki Tokola
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Department of Pathology, Institute of Diagnostics, University of Oulu, Oulu, Finland
| | - Jani Aro
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Harri Pennanen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Hanna Säkkinen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
| | - Heikki Ruskoaho
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Jaana Rysä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
13
|
Huusko J, Lottonen L, Merentie M, Gurzeler E, Anisimov A, Miyanohara A, Alitalo K, Tavi P, Ylä-Herttuala S. AAV9-mediated VEGF-B gene transfer improves systolic function in progressive left ventricular hypertrophy. Mol Ther 2012; 20:2212-21. [PMID: 23089731 DOI: 10.1038/mt.2012.145] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mechanisms of the transition from compensatory hypertrophy to heart failure are poorly understood and the roles of vascular endothelial growth factors (VEGFs) in this process have not been fully clarified. We determined the expression profile of VEGFs and relevant receptors during the progression of left ventricular hypertrophy (LVH). C57BL mice were exposed to transversal aortic constriction (TAC) and the outcome was studied at different time points (1 day, 2, 4, and 10 weeks). A clear compensatory phase (2 weeks after TAC) was seen with following heart failure (4 weeks after TAC). Interestingly, VEGF-C and VEGF-D as well as VEGF receptor-3 (VEGFR-3) were upregulated in the compensatory hypertrophy and VEGF-B was downregulated in the heart failure. After treatment with adeno-associated virus serotype 9 (AAV9)-VEGF-B(186) gene therapy in the compensatory phase for 4 weeks the function of the heart was preserved due to angiogenesis, inhibition of apoptosis, and promotion of cardiomyocyte proliferation. Also, the genetic programming towards fetal gene expression, a known phenomenon in heart failure, was partly reversed in AAV9-VEGF-B(186)-treated mice. We conclude that VEGF-C and VEGF-D are associated with the compensatory LVH and that AAV9-VEGF-B(186) gene transfer can rescue the function of the failing heart and postpone the transition towards heart failure.
Collapse
Affiliation(s)
- Jenni Huusko
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kameda Y, Hasegawa H, Kubota A, Tadokoro H, Kobayashi Y, Komuro I, Takano H. Effects of Pitavastatin on Pressure Overload-Induced Heart Failure in Mice. Circ J 2012; 76:1159-68. [DOI: 10.1253/circj.cj-11-1114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yoshihito Kameda
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine
| | - Hiroshi Hasegawa
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine
| | - Akihiko Kubota
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine
| | - Hiroyuki Tadokoro
- Department of Bio-Medical Engineering, Tokai University School of High-Technology for Human Welfare
| | - Yoshio Kobayashi
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine
| | - Issei Komuro
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Hiroyuki Takano
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
15
|
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
|
16
|
Razani B, Zhang H, Schulze PC, Schilling JD, Verbsky J, Lodhi IJ, Topkara VK, Feng C, Coleman T, Kovacs A, Kelly DP, Saffitz JE, Dorn GW, Nichols CG, Semenkovich CF. Fatty acid synthase modulates homeostatic responses to myocardial stress. J Biol Chem 2011; 286:30949-30961. [PMID: 21757749 DOI: 10.1074/jbc.m111.230508] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Fatty acid synthase (FAS) promotes energy storage through de novo lipogenesis and participates in signaling by the nuclear receptor PPARα in noncardiac tissues. To determine if de novo lipogenesis is relevant to cardiac physiology, we generated and characterized FAS knockout in the myocardium (FASKard) mice. FASKard mice develop normally, manifest normal resting heart function, and have normal cardiac PPARα signaling as well as fatty acid oxidation. However, they decompensate with stress. Most die within 1 h of transverse aortic constriction, probably due to arrhythmia. Voltage clamp measurements of FASKard cardiomyocytes show hyperactivation of L-type calcium channel current that could not be reversed with palmitate supplementation. Of the classic regulators of this current, Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) but not protein kinase A signaling is activated in FASKard hearts, and knockdown of FAS in cultured cells activates CaMKII. In addition to being intolerant of the stress of acute pressure, FASKard hearts were also intolerant of the stress of aging, reflected as persistent CaMKII hyperactivation, progression to dilatation, and premature death by ∼1 year of age. CaMKII signaling appears to be pathogenic in FASKard hearts because inhibition of its signaling in vivo rescues mice from early mortality after transverse aortic constriction. FAS was also increased in two mechanistically distinct mouse models of heart failure and in the hearts of humans with end stage cardiomyopathy. These data implicate a novel relationship between FAS and calcium signaling in the heart and suggest that FAS induction in stressed myocardium represents a compensatory response to protect cardiomyocytes from pathological calcium flux.
Collapse
Affiliation(s)
- Babak Razani
- Divisions of Endocrinology, Metabolism, and Lipid Research, Washington University, St. Louis, Missouri 63110; Cardiology, Washington University, St. Louis, Missouri 63110
| | - Haixia Zhang
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | | | | | - John Verbsky
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | - Irfan J Lodhi
- Divisions of Endocrinology, Metabolism, and Lipid Research, Washington University, St. Louis, Missouri 63110
| | - Veli K Topkara
- Cardiology, Washington University, St. Louis, Missouri 63110
| | - Chu Feng
- Divisions of Endocrinology, Metabolism, and Lipid Research, Washington University, St. Louis, Missouri 63110
| | - Trey Coleman
- Divisions of Endocrinology, Metabolism, and Lipid Research, Washington University, St. Louis, Missouri 63110
| | - Attila Kovacs
- Cardiology, Washington University, St. Louis, Missouri 63110
| | - Daniel P Kelly
- Sanford-Burnham Medical Research Institute, Orlando, Florida 32827
| | - Jeffrey E Saffitz
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
| | - Gerald W Dorn
- Center for Pharmacogenomics, Washington University, St. Louis, Missouri 63110
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | - Clay F Semenkovich
- Divisions of Endocrinology, Metabolism, and Lipid Research, Washington University, St. Louis, Missouri 63110; Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110.
| |
Collapse
|
17
|
Wu X, Sun Z, Foskett A, Trzeciakowski JP, Meininger GA, Muthuchamy M. Cardiomyocyte contractile status is associated with differences in fibronectin and integrin interactions. Am J Physiol Heart Circ Physiol 2010; 298:H2071-81. [PMID: 20382852 DOI: 10.1152/ajpheart.01156.2009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Integrins link the extracellular matrix (ECM) with the intracellular cytoskeleton and other cell adhesion-associated signaling proteins to function as mechanotransducers. However, direct quantitative measurements of the cardiomyocyte mechanical state and its relationship to the interactions between specific ECM proteins and integrins are lacking. The purpose of this study was to characterize the interactions between the ECM protein fibronectin (FN) and integrins in cardiomyocytes and to test the hypothesis that these interactions would vary during contraction and relaxation states in cardiomyocytes. Using atomic force microscopy, we quantified the unbinding force (adhesion force) and adhesion probability between integrins and FN and correlated these measurements with the contractile state as indexed by cell stiffness on freshly isolated mouse cardiomyocytes. Experiments were performed in normal physiological (control), high-K(+) (tonically contracted), or low-Ca(2+) (fully relaxed) solutions. Under control conditions, the initial peak of adhesion force between FN and myocyte alpha(3)beta(1)- and/or alpha(5)beta(1)-integrins was 39.6 +/- 1.3 pN. The binding specificity between FN and alpha(3)beta(1)- and alpha(5)beta(1)-integrins was verified by using monoclonal antibodies against alpha(3)-, alpha(5)-, alpha(3) + alpha(5)-, or beta(1)-integrin subunits, which inhibited binding by 48%, 65%, 70%, or 75%, respectively. Cytochalasin D or 2,3-butanedione monoxime (BDM), to disrupt the actin cytoskeleton or block myofilament function, respectively, significantly decreased the cell stiffness; however, the adhesion force and binding probability were not altered. Tonic contraction with high-K(+) solution increased total cell adhesion (1.2-fold) and cell stiffness (27.5-fold) compared with fully relaxed cells with low-Ca(2+) solution. However, it could be partially prevented by high-K(+) bath solution containing BDM, which suppresses contraction by inhibiting the actin-myosin interactions. Thus, our results demonstrate that integrin binding to FN is modulated by the contractile state of cardiac myocytes.
Collapse
Affiliation(s)
- Xin Wu
- Dept. of Systems Biology and Translational Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX 77843-1114, USA
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Magnetic nanoparticles can be coated with specific ligands that enable them to bind to receptors on a cell's surface. When a magnetic field is applied, it pulls on the particles so that they deliver nanoscale forces at the ligand-receptor bond. It has been observed that mechanical stimulation in this manner can activate cellular signaling pathways that are known as mechanotransduction pathways. Integrin receptors, stretch-activated ion channels, focal adhesions, and the cytoskeleton are key players in activating these pathways, but there is still much we do not know about how these mechanosensors work. Current evidence indicates that applied forces at these structures can activate Ca(2+) signaling, Src family protein kinase, MAPK, and RhoGTPase pathways. The techniques of magnetic twisting and magnetic tweezers, which use magnetic particles to apply forces to cells, afford a fine degree of control over how cells are stimulated and hold much promise in elucidating the fundamentals of mechanotransduction. The particles are generally not harmful to cellular health, and their nanoscale dimensions make them advantageous for probing a cell's molecular-scale sensory structures. This review highlights the basic aspects of magnetic nanoparticles, magnetic particle techniques and the structures and pathways that are involved in mechanotransduction.
Collapse
Affiliation(s)
- Nathan J Sniadecki
- Department of Mechanical Engineering, Box 352600 University of Washington, Seattle, Washington 98195, USA.
| |
Collapse
|
19
|
Colak D, Kaya N, Al-Zahrani J, Al Bakheet A, Muiya P, Andres E, Quackenbush J, Dzimiri N. Left ventricular global transcriptional profiling in human end-stage dilated cardiomyopathy. Genomics 2009; 94:20-31. [PMID: 19332114 PMCID: PMC4152850 DOI: 10.1016/j.ygeno.2009.03.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 02/17/2009] [Accepted: 03/17/2009] [Indexed: 02/07/2023]
Abstract
We employed ABI high-density oligonucleotide microarrays containing 31,700 sixty-mer probes (representing 27,868 annotated human genes) to determine differential gene expression in idiopathic dilated cardiomyopathy (DCM). We identified 626 up-regulated and 636 down-regulated genes in DCM compared to controls. Most significant changes occurred in the tricarboxylic acid cycle, angiogenesis, and apoptotic signaling pathways, among which 32 apoptosis- and 13 MAPK activity-related genes were altered. Inorganic cation transporter, catalytic activities, energy metabolism and electron transport-related processes were among the most critically influenced pathways. Among the up-regulated genes were HTRA1 (6.9-fold), PDCD8(AIFM1) (5.2) and PRDX2 (4.4) and the down-regulated genes were NR4A2 (4.8), MX1 (4.3), LGALS9 (4), IFNA13 (4), UNC5D (3.6) and HDAC2 (3) (p<0.05), all of which have no clearly defined cardiac-related function yet. Gene ontology and enrichment analysis also revealed significant alterations in mitochondrial oxidative phosphorylation, metabolism and Alzheimer's disease pathways. Concordance was also confirmed for a significant number of genes and pathways in an independent validation microarray dataset. Furthermore, verification by real-time RT-PCR showed a high degree of consistency with the microarray results. Our data demonstrate an association of DCM with alterations in various cellular events and multiple yet undeciphered genes that may contribute to heart muscle disease pathways.
Collapse
Affiliation(s)
- Dilek Colak
- Department of Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Namik Kaya
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Jawaher Al-Zahrani
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Albandary Al Bakheet
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Paul Muiya
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Editha Andres
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - John Quackenbush
- Department of Biostatistics and Computational Biology; Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nduna Dzimiri
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| |
Collapse
|
20
|
Schwartz MA, DeSimone DW. Cell adhesion receptors in mechanotransduction. Curr Opin Cell Biol 2008; 20:551-6. [PMID: 18583124 DOI: 10.1016/j.ceb.2008.05.005] [Citation(s) in RCA: 322] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 05/13/2008] [Accepted: 05/19/2008] [Indexed: 12/14/2022]
Abstract
Integrins and cadherins are tri-functional: they bind ligands on other cells or in the extracellular matrix, connect to the cytoskeleton inside the cell, and regulate intracellular signaling pathways. These adhesion receptors therefore transmit mechanical stresses and are well positioned to mediate mechanotransduction. Studies of cultured cells have shown that both integrin- and cadherin-mediated adhesion are intrinsically mechanosensitive. Strengthening of adhesions in response to mechanical stimulation may be a central mechanism for mechanotransduction. Studies of developing organisms suggest that these mechanisms contribute to tissue level responses to tension and compression, thereby linking morphogenetic movements to cell fate decisions.
Collapse
Affiliation(s)
- Martin A Schwartz
- Department of Microbiology and Biomedical Engineering, Cardiovascular Research Center and Mellon Urological Cancer Research Institute, University of Virginia Charlottesville, VA 22908, USA.
| | | |
Collapse
|
21
|
Ronkainen JJ, Vuolteenaho O, Tavi P. Calcium-calmodulin kinase II is the common factor in calcium-dependent cardiac expression and secretion of A- and B-type natriuretic peptides. Endocrinology 2007; 148:2815-20. [PMID: 17332063 DOI: 10.1210/en.2006-1676] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peptides derived from the precursor of A- and B-type natriuretic peptides (ANP and BNP) are powerful clinical markers of cardiac hypertrophy and dysfunction. It is known that many stimuli affecting the intracellular calcium concentration also induce ANP and BNP secretion. It was our intention to study the mechanisms by which calcium regulates the secretion of ANP and BNP. The effects of pacing and calcium-calmodulin kinase II activity on natriuretic peptide secretion were studied in isolated perfused rat atria and cultured rat neonatal cardiomyocytes. In isolated rat atrium pacing induced an increase in diastolic, systolic, and averaged intracellular free calcium concentration and a frequency-dependent increase in the secretion of both ANP and BNP. The molar ratio of the secreted natriuretic peptides (ANP to BNP) remained nearly constant ( approximately 1000) at all the pacing frequencies tested (1, 3, 6, and 8 Hz). Calmodulin kinase II inhibitor KN-93 (3 mum) did not affect intracellular free calcium concentration but showed a frequency-dependent inhibitory effect on ANP and BNP secretion without a change in ANP to BNP ratio. In the neonatal cardiomyocytes, KN-93 (3 mum) suppressed the secretion and gene expression of both ANP and BNP. Overexpression of constitutively active (T286D) or nuclear (delta(B)) calcium-calmodulin kinase II induced an increase in ANP and BNP gene expression. The results indicate that the calcium-dependent secretion and gene expression of A- and B-type natriuretic peptides are similarly regulated by calmodulin kinase II-dependent mechanisms. This is a plausible mechanism contributing to exercise-induced natriuretic peptide secretion and the augmented secretion in heart dysfunction due to impaired calcium handling.
Collapse
Affiliation(s)
- Jarkko J Ronkainen
- University of Oulu, Department of Physiology and Biocenter Oulu, University of Oulu, 90014 Oulu, Finland
| | | | | |
Collapse
|
22
|
|
23
|
Hasegawa H, Takano H, Kohro T, Ueda K, Niitsuma Y, Aburatani H, Komuro I. Amelioration of hypertensive heart failure by amlodipine may occur via antioxidative effects. Hypertens Res 2007; 29:719-29. [PMID: 17249528 DOI: 10.1291/hypres.29.719] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although recent clinical studies have suggested that long-acting calcium channel blockers (CCBs) have beneficial effects on heart failure, the precise mechanism is unknown. In this study, Dahl salt-sensitive rats fed a high salt diet were treated with the long-acting CCB amlodipine, the low-molecular-weight membrane permeable superoxide dismutase mimetic 4-hydroxy-2,2,6,6-tetramethyl piperidinoxyl (Tempol), or saline from 11 weeks after birth. The cardiac geometry and function, and gene expression profiles were determined at 17 weeks. Dahl salt-sensitive rats fed a high salt diet followed by saline as a non-treatment control (HS group) showed a marked increase in blood pressure and developed concentric hypertrophy at 11 weeks, followed by left ventricular (LV) dilation and congestive heart failure by 17 weeks. The treatment with amlodipine (AMLO group) or Tempol (TEMP group) significantly inhibited the development of LV hypertrophy and cardiac dysfunction. Analysis using an Affymetrix GeneChip U34 revealed that the expression levels of 195 genes were changed by the treatment with amlodipine. Among these 195 genes, 110 genes were increased in HS rats and decreased in AMLO rats. And of these 110 genes, 54 genes were also decreased in TEMP rats. In contrast, 85 genes were decreased in HS rats and increased in AMLO rats. Of these 85 genes, 38 genes were also increased in TEMP rats. Approximately 48% of the genes were changed in similar fashion in AMLO and TEMP rats, suggesting that amlodipine shows beneficial effects on heart failure mainly via antioxidative mechanisms.
Collapse
Affiliation(s)
- Hiroshi Hasegawa
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Heymans S, Lupu F, Terclavers S, Vanwetswinkel B, Herbert JM, Baker A, Collen D, Carmeliet P, Moons L. Loss or inhibition of uPA or MMP-9 attenuates LV remodeling and dysfunction after acute pressure overload in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:15-25. [PMID: 15631996 PMCID: PMC1602291 DOI: 10.1016/s0002-9440(10)62228-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Left ventricular (LV) hypertrophy is a natural response of the heart to increased pressure loading, but accompanying fibrosis and dilatation may result in irreversible life-threatening heart failure. Matrix metalloproteinases (MMPs) have been invoked in various cardiac diseases, however, direct genetic evidence for a role of the plasminogen activator (PA) and MMP systems in pressure overload-induced LV hypertrophy and in heart failure is lacking. Therefore, the consequences of transverse aortic banding (TAB) were analyzed in mice lacking tissue-type PA (t-PA(-/-)), urokinase-type PA (u-PA(-/-)), or gelatinase-B (MMP-9(-/-)), and in wild-type (WT) mice after adenoviral gene transfer of the PA-inhibitor PAI-1 or the MMP-inhibitor TIMP-1. TAB elevated LV pressure comparably in all genotypes. In WT and t-PA(-/-) mice, cardiomyocyte hypertrophy was associated with myocardial fibrosis, LV dilatation and dysfunction, and pump failure after 7 weeks. In contrast, in u-PA(-/-) mice or in WT mice after PAI-1- and TIMP-1-gene transfer, cardiomyocyte hypertrophy was moderate and only minimally associated with cardiac fibrosis and LV dilatation, resulting in better preservation of pump function. Deficiency of MMP-9 had an intermediate effect. These findings suggest that the use of u-PA- or MMP-inhibitors might preserve cardiac pump function in LV pressure overloading.
Collapse
Affiliation(s)
- Stephane Heymans
- Center for Transgene Technology and Gene Therapy, Flanders Interuniversity Institute, Herestraat 49, B-3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Syntichaki P, Tavernarakis N. Genetic Models of Mechanotransduction: The NematodeCaenorhabditis elegans. Physiol Rev 2004; 84:1097-153. [PMID: 15383649 DOI: 10.1152/physrev.00043.2003] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mechanotransduction, the conversion of a mechanical stimulus into a biological response, constitutes the basis for a plethora of fundamental biological processes such as the senses of touch, balance, and hearing and contributes critically to development and homeostasis in all organisms. Despite this profound importance in biology, we know remarkably little about how mechanical input forces delivered to a cell are interpreted to an extensive repertoire of output physiological responses. Recent, elegant genetic and electrophysiological studies have shown that specialized macromolecular complexes, encompassing mechanically gated ion channels, play a central role in the transformation of mechanical forces into a cellular signal, which takes place in mechanosensory organs of diverse organisms. These complexes are highly efficient sensors, closely entangled with their surrounding environment. Such association appears essential for proper channel gating and provides proximity of the mechanosensory apparatus to the source of triggering mechanical energy. Genetic and molecular evidence collected in model organisms such as the nematode worm Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and the mouse highlight two distinct classes of mechanically gated ion channels: the degenerin (DEG)/epithelial Na+channel (ENaC) family and the transient receptor potential (TRP) family of ion channels. In addition to the core channel proteins, several other potentially interacting molecules have in some cases been identified, which are likely parts of the mechanotransducing apparatus. Based on cumulative data, a model of the sensory mechanotransducer has emerged that encompasses our current understanding of the process and fulfills the structural requirements dictated by its dedicated function. It remains to be seen how general this model is and whether it will withstand the impiteous test of time.
Collapse
Affiliation(s)
- Popi Syntichaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Vassilika Vouton, PO Box 1527, Heraklion 71110, Crete, Greece
| | | |
Collapse
|
27
|
Abstract
Cardiac myocytes react to diverse mechanical demands with a multitude of transient and long-term responses to normalize the cellular mechanical environment. Several stretch-activated signaling pathways have been identified, most prominently guanine nucleotide binding proteins (G-proteins), mitogen-activated protein kinases (MAPK), Janus-associated kinase/signal transducers and activators of transcription (JAK/STAT), protein kinase C (PKC), calcineurin, intracellular calcium regulation, and several autocrine and paracrine factors. Multiple levels of crosstalk exist between pathways. The cellular response to changes in the mechanical environment can lead to cardiac myocyte hypertrophy, cellular growth that can be accompanied by pathological myocyte dysfunction, and tissue fibrosis. Several candidates for the primary mechanosensor in cardiac myocytes have been identified, ranging from stretch-activated ion channels in the membrane to yet-unknown mechanosensitive mechanisms in the nucleus. New and refined experimental techniques will exploit advances in molecular biology and biological imaging to study mechanotransduction in isolated cells and genetically engineered mice to explore the function of individual proteins.
Collapse
Affiliation(s)
- Jan Lammerding
- Biological Engineering Division, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
28
|
Huang H, Kamm RD, Lee RT. Cell mechanics and mechanotransduction: pathways, probes, and physiology. Am J Physiol Cell Physiol 2004; 287:C1-11. [PMID: 15189819 DOI: 10.1152/ajpcell.00559.2003] [Citation(s) in RCA: 344] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cells face not only a complex biochemical environment but also a diverse biomechanical environment. How cells respond to variations in mechanical forces is critical in homeostasis and many diseases. The mechanisms by which mechanical forces lead to eventual biochemical and molecular responses remain undefined, and unraveling this mystery will undoubtedly provide new insight into strengthening bone, growing cartilage, improving cardiac contractility, and constructing tissues for artificial organs. In this article we review the physical bases underlying the mechanotransduction process, techniques used to apply controlled mechanical stresses on living cells and tissues to probe mechanotransduction, and some of the important lessons that we are learning from mechanical stimulation of cells with precisely controlled forces.
Collapse
Affiliation(s)
- Hayden Huang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139, USA.
| | | | | |
Collapse
|
29
|
Ruzicska E, Foldes G, Lako-Futo Z, Sarman B, Wellmann J, Szenasi G, Tulassay Z, Ruskoaho H, Toth M, Somogyi A. Cardiac gene expression of natriuretic substances is altered in streptozotocin-induced diabetes during angiotensin II-induced pressure overload. J Hypertens 2004; 22:1191-200. [PMID: 15167455 DOI: 10.1097/00004872-200406000-00021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To gain insight into the cardiac adaptive mechanisms in diabetes, we studied whether angiotensin II (Ang II) alters expression of the atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP) and adrenomedullin (AM) genes in the left ventricle of the diabetic rat heart. METHODS Diabetes was induced by streptozotocin (STZ; 60 mg/kg body weight intravenously). During the last 24 h of 2.5 or 7 weeks of treatment of male Wistar rats with STZ or vehicle, Ang II (33 microg/kg per h) was administered via osmotic minipumps. RESULTS Diabetes was associated with an increased left ventricular weight to body weight (LV/BW) ratio, an index of left ventricular hypertrophy, at week 7 but not at week 2.5, and with increased ANP mRNA content at 2.5 weeks, but not with altered expression of the AM and BNP genes. Mean arterial pressure and LV/BW ratio were increased by Ang II in all groups except in the 7-week diabetic group. Levels of ANP mRNA were increased fourfold (P < 0.001) and threefold (P < 0.05) by Ang II at 2.5 and 7 weeks in control animals, respectively, and 11-fold (P < 0.001) and sevenfold (P < 0.001) at 2.5 and 7 weeks in diabetic animals, respectively. Ang II increased ventricular concentrations of BNP mRNA in control and diabetic animals at 2.5 weeks (1.3-fold, P < 0.001; and 1.6-fold, P < 0.001) and at 7 weeks (1.3-fold, P < 0.05; and 1.8-fold, P < 0.001), respectively. Left ventricular levels of adrenomedullin mRNA were increased by treatment with Ang II for 24 h in 2.5-week diabetic animals. CONCLUSION Ang II markedly increased the levels of natriuretic peptide mRNAs in the left ventricle of normal and diabetic rat hearts, whereas it increased adrenomedullin mRNA levels only in 2.5-week diabetic rats and failed to cause hypertension in 7-week diabetic rats. Left ventricular levels of ANP and BNP mRNA were increased by Ang II in diabetic animals more than the additive effects of diabetes and Ang II alone, showing that Ang II induced an amplified response with respect to cardiac concentrations of ANP and BNP in diabetes.
Collapse
Affiliation(s)
- Eva Ruzicska
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tavi P, Pikkarainen S, Ronkainen J, Niemelä P, Ilves M, Weckström M, Vuolteenaho O, Bruton J, Westerblad H, Ruskoaho H. Pacing-induced calcineurin activation controls cardiac Ca2+ signalling and gene expression. J Physiol 2004; 554:309-20. [PMID: 14565991 PMCID: PMC1664772 DOI: 10.1113/jphysiol.2003.053579] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2003] [Accepted: 10/16/2003] [Indexed: 12/19/2022] Open
Abstract
Calcineurin, a Ca(2+)-calmodulin-dependent protein phosphatase (PP2B) is one of the links between Ca(2+) signals and regulation of gene transcription in cardiac muscle. We studied the Ca(2+) signal specificity of calcineurin activation experimentally and with modelling. In the rat atrial preparation, an increase in pacing frequency increased nuclear activity of the calcineurin-sensitive transcription factor, nuclear factor of activated T-cells (NFAT), 2-fold in a cyclosporin A (CsA)-sensitive manner. In line with this, modelling results predicted that the frequency of cardiac Ca(2+) transients encodes the stimulus for calcineurin activation. We further observed experimentally that calcineurin inhibition by CsA modulated Ca(2+) release in a Ca(2+)-dependent manner. CsA had no effect on [Ca(2+)](i) at a pacing frequency of 1 Hz but it significantly suppressed the amplitude of Ca(2+) transients, systolic [Ca(2+)](i) and time averaged [Ca(2+)](i) at 6 Hz. Calcineurin had a differential role in the expression of immediate-early genes B-type natriuretic peptide (BNP) and c-fos. CsA inhibited the pacing-induced BNP gene expression, whereas pacing alone had no effect on the expression of c-fos. However, in the presence of CsA, c-fos mRNA levels were significantly augmented by increased pacing frequency. These results show that frequency-dependent calcineurin activation has a specific role in [Ca(2+)](i) regulation and gene expression, constantly recruited by varying cardiac Ca(2+) signals.
Collapse
Affiliation(s)
- Pasi Tavi
- Department of Physiology, Biocentre Oulu, University of Oulu, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Piuhola J, Mäkinen M, Szokodi I, Ruskoaho H. Dual role of endothelin-1 via ETA and ETB receptors in regulation of cardiac contractile function in mice. Am J Physiol Heart Circ Physiol 2003; 285:H112-8. [PMID: 12609819 DOI: 10.1152/ajpheart.00480.2002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
An increase in coronary perfusion pressure leads to increased cardiac contractility, a phenomenon known as the Gregg effect. Exogenous endothelin (ET)-1 exerts a positive inotropic effect; however, the role of endogenous ET-1 in the contractile response to elevated load is unknown. We characterized here the role of ETA and ETB receptors in regulation of contractility in isolated, perfused mouse hearts subjected to increased coronary flow. Elevation of coronary flow from 2 to 5 ml/min resulted in 80 +/- 10% increase in contractile force (P < 0.001). BQ-788 (ETB receptor antagonist) augmented the load-induced contractile response by 35% (P < 0.05), whereas bosentan (ETA/B receptor antagonist) and BQ-123 (ETA receptor antagonist) attenuated it by 34% and 56%, respectively (P < 0.05). CV-11974 (ANG II type 1 receptor antagonist) did not modify the increase in contractility. These results show that endogenous ET-1 is a key mediator of the Gregg effect in mouse hearts. Moreover, ET-1 has a dual role in the regulation of cardiac contractility: ETA receptor-mediated increase in contractile force is suppressed by ETB receptors.
Collapse
Affiliation(s)
- Jarkko Piuhola
- Department of Pharmacology and Toxicology, University of Oulu, 90014 Oulu, Finland
| | | | | | | |
Collapse
|
32
|
Affiliation(s)
- John D Baxter
- The Diabetes Center, University of California, San Francisco, California 94122, USA.
| | | | | |
Collapse
|
33
|
Latimer DC, Roth BJ, Parker KK. Analytical model for predicting mechanotransduction effects in engineered cardiac tissue. TISSUE ENGINEERING 2003; 9:283-9. [PMID: 12740090 DOI: 10.1089/107632703764664747] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Mechanochemical and mechanoelectrical signaling is imperative for cardiac organogenesis and underlies pathophysiological events. New techniques for engineering cardiac tissue allow unprecedented means of modeling these phenomena in vitro. However, experimental design is often hampered by a lack of models that can be adapted to the ideal conditions these methods allow. To address these deficiencies, we developed a mathematical model to calculate the distribution of stress and strain in fibrous cardiac tissue. The fluid-fiber-collagen model characterizes the mechanical behavior of cardiac tissue and is solved analytically for the distributions of stress and strain along the myocardial fibers. An example application of the model is presented: modeling the distribution of strains in the vicinity of an ischemic region. The ischemic region is stretched during systole, as has been shown in previous one-dimensional models. Our model predicts a complex distribution of stretch in the border zone surrounding the ischemic region and in nonischemic regions surrounding the border zone. These strain patterns may predict patterns of mechanochemical coupling that results in localized fibrosis, altered gene expression, or the mechanoelectrical signaling events that potentiate cardiac arrhythmias.
Collapse
Affiliation(s)
- David C Latimer
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee 48309, USA
| | | | | |
Collapse
|
34
|
Piuhola J, Szokodi I, Kinnunen P, Ilves M, deChâtel R, Vuolteenaho O, Ruskoaho H. Endothelin-1 contributes to the Frank-Starling response in hypertrophic rat hearts. Hypertension 2003; 41:93-8. [PMID: 12511536 DOI: 10.1161/01.hyp.0000050929.96979.ec] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Endothelin-1 is involved in mechanical load-induced cardiac growth processes; it also has effects on contractility. The interaction of endothelin-1 and the Frank-Starling response is unknown. The present study aimed to characterize the role of endothelin-1 in the regulation of the Frank-Starling response, one of the major mechanisms regulating cardiac contractile force, in both normal and hypertrophied hearts. Nontransgenic rat hearts and hypertrophic hearts of hypertensive double transgenic rats harboring human angiotensinogen and renin genes were studied in a Langendorff isolated heart setup with a liquid-filled balloon inside the left ventricle used to measure contractile parameters. The rats were studied at compensated phase, before showing any signs of heart failure. Compensated hypertrophy in double transgenic rat hearts resulted in improved contractility at a given level of preload when compared with nontransgenic rat hearts. Hearts of both rat lines showed preserved Frank-Starling responses, that is, increased contractile function in response to increased end-diastolic pressure. The mixed endothelin A/B receptor antagonist bosentan attenuated the Frank-Starling response by 53% (P<0.01) in the double transgenic hearts but not in nontransgenic hearts. The diastolic parameters remained unaffected. The left ventricles of the double transgenic rat hearts showed an 82% higher level of endothelin type A receptor mRNA and a 25% higher level of immunoreactive endothelin-1 compared with nontransgenic rat hearts. The type 1 angiotensin II receptor antagonist CV-11974 had no significant effect on contractile function in response to load in either strain. These results show that endogenous endothelin-1 contributes to the Frank-Starling response in hypertrophied rat hearts by affecting systolic performance.
Collapse
MESH Headings
- Angiotensin II/physiology
- Angiotensin Receptor Antagonists
- Angiotensinogen/genetics
- Animals
- Animals, Genetically Modified
- Benzimidazoles/pharmacology
- Biphenyl Compounds
- Bosentan
- Endothelin Receptor Antagonists
- Endothelin-1/physiology
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Humans
- Hypertension/complications
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Myocardial Contraction
- Organ Culture Techniques
- RNA, Messenger/biosynthesis
- Rats
- Rats, Inbred SHR
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1
- Renin/genetics
- Stress, Mechanical
- Sulfonamides/pharmacology
- Tetrazoles/pharmacology
Collapse
Affiliation(s)
- Jarkko Piuhola
- Department of Pharmacology and Toxicology, Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | |
Collapse
|
35
|
Kerkelä R, Ilves M, Pikkarainen S, Tokola H, Ronkainen J, Vuolteenaho O, Leppäluoto J, Ruskoaho H. Identification of PKCalpha isoform-specific effects in cardiac myocytes using antisense phosphorothioate oligonucleotides. Mol Pharmacol 2002; 62:1482-91. [PMID: 12435817 DOI: 10.1124/mol.62.6.1482] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Members of the mammalian protein kinase C (PKC) superfamily play key regulatory roles in multiple cellular processes. In the heart, PKC signaling is involved in hypertrophic agonist-induced gene expression and hypertrophic growth. To investigate the specific function of PKC signaling in regulating cardiomyocyte growth, we used antisense oligonucleotides to inhibit PKC alpha, the major isozyme present in the neonatal heart. Transfection of cultured neonatal cardiomyocytes with antisense PKCalpha oligonucleotides resulted in a marked reduction in both PKCalpha mRNA and protein levels. PKCalpha antisense treatment also reduced phenylephrine (PE)-induced PKC activity and perinuclear translocation of PKCalpha. Antisense inhibition of PKCalpha led to reduction of PE-induced increase in skeletal alpha-actin mRNA levels and atrial natriuretic peptide (ANP) secretion but had no significant effects on PE-induced beta-myosin heavy chain, ANP, or B-type natriuretic peptide (BNP) gene expression. On the other hand, antisense PKCalpha treatment attenuated endothelin-1-induced increase in ANP and BNP peptide secretion, whereas endothelin-1-induced gene expression of ANP and BNP remained unchanged. The hypertrophic agonist-induced growth of cardiomyocytes, characterized by increased [(3)H]leucine incorporation, was not affected with antisense PKCalpha treatment. Furthermore, we found that PE-induced increase in extracellular signal-regulated kinase (ERK) activity was partially inhibited by antisense PKCalpha treatment, implicating ERK as a downstream mediator for PKCalpha signaling. These results indicate that PKCalpha isozyme is involved in hypertrophic signaling in cardiomyocytes and provide novel strategies for future studies to identify other cellular targets controlled selectively by PKCalpha or other PKC isozymes.
Collapse
Affiliation(s)
- Risto Kerkelä
- Department of Pharmacology and Toxicology, Biocenter Oulu, University of Oulu, Finland
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Persoon-Rothert M, van der Wees KGC, van der Laarse A. Mechanical overload-induced apoptosis: a study in cultured neonatal ventricular myocytes and fibroblasts. Mol Cell Biochem 2002; 241:115-24. [PMID: 12482033 DOI: 10.1023/a:1020860209333] [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] [Indexed: 11/12/2022]
Abstract
Apoptosis of cardiac myocytes has been implicated in cardiac dysfunction due to chronic hemodynamic overload. Reports on the role of apoptosis in the transition from hypertrophy to decompensated heart failure are not unequivocal. In this study we analysed the direct relationship between mechanical overload and induction of apoptosis in an in vitro model of cultured heart cells. Cyclic mechanical stretch was applied to cultured neonatal rat ventricular myocytes and fibroblasts. Several indicators of apoptosis were examined, such as morphological features, caspase-3 activity and DNA fragmentation. Mechanical strain did not induce any significant change in these parameters as compared to non-stretched myocytes or fibroblasts. However, administration of staurosporine, a known inducer of apoptosis, induced massive apoptosis in myocytes as well as fibroblasts. We conclude that this in vitro cell model system lacks a direct link between mechanical stretch and apoptosis. The three-dimensional structure-function relationship of myocardial tissue in the intact heart may elicit stretch-induced molecular signaling cascades in a much more complex way than in monolayer cultures of cardiac cells.
Collapse
|
37
|
Abstract
Biomechanical signaling is a complex interaction of both intracellular and extracellular components. Both passive and active components are involved in the extracellular environment to signal through specific receptors to multiple signaling pathways. This review provides an overview of extracellular matrix, specific receptors, and signaling pathways for biomechanical stimulation in cardiac hypertrophy.
Collapse
Affiliation(s)
- Mark A Sussman
- Children's Hospital and Research Foundation, Division of Molecular Cardiovascular Biology, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
38
|
Szokodi I, Tavi P, Földes G, Voutilainen-Myllylä S, Ilves M, Tokola H, Pikkarainen S, Piuhola J, Rysä J, Tóth M, Ruskoaho H. Apelin, the novel endogenous ligand of the orphan receptor APJ, regulates cardiac contractility. Circ Res 2002; 91:434-40. [PMID: 12215493 DOI: 10.1161/01.res.0000033522.37861.69] [Citation(s) in RCA: 456] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The orphan receptor APJ and its recently identified endogenous ligand, apelin, exhibit high levels of mRNA expression in the heart. However, the functional importance of apelin in the cardiovascular system is not known. In isolated perfused rat hearts, infusion of apelin (0.01 to 10 nmol/L) induced a dose-dependent positive inotropic effect (EC50: 33.1+/-1.5 pmol/L). Moreover, preload-induced increase in dP/dt(max) was significantly augmented (P<0.05) in the presence of apelin. Inhibition of phospholipase C (PLC) with U-73122 and suppression of protein kinase C (PKC) with staurosporine and GF-109203X markedly attenuated the apelin-induced inotropic effect (P<0.001). In addition, zoniporide, a selective inhibitor of Na+-H+ exchange (NHE) isoform-1, and KB-R7943, a potent inhibitor of the reverse mode Na+-Ca2+ exchange (NCX), significantly suppressed the response to apelin (P<0.001). Perforated patch-clamp recordings showed that apelin did not modulate L-type Ca2+ current or voltage-activated K+ currents in isolated adult rat ventricular myocytes. Apelin mRNA was markedly downregulated in cultured neonatal rat ventricular myocytes subjected to mechanical stretch and in vivo in two models of chronic ventricular pressure overload. The present study provides the first evidence for the physiological significance of apelin in the heart. Our results show that apelin is one of the most potent endogenous positive inotropic substances yet identified and that the inotropic response to apelin may involve activation of PLC, PKC, and sarcolemmal NHE and NCX.
Collapse
MESH Headings
- Adrenomedullin
- Animals
- Animals, Genetically Modified
- Apelin
- Apelin Receptors
- Calcium Channels/physiology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Carrier Proteins/pharmacology
- Dose-Response Relationship, Drug
- Endothelin-1/pharmacology
- Gene Expression Regulation
- Heart Ventricles/cytology
- Heart Ventricles/drug effects
- In Vitro Techniques
- Intercellular Signaling Peptides and Proteins
- Isoproterenol/pharmacology
- Ligands
- Male
- Membrane Potentials/drug effects
- Myocardial Contraction/drug effects
- Peptides/pharmacology
- Potassium Channels/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Rats, Sprague-Dawley
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Receptors, G-Protein-Coupled
- Sodium-Calcium Exchanger/metabolism
- Sodium-Hydrogen Exchangers/metabolism
- Stress, Mechanical
- Time Factors
- Ventricular Function
Collapse
Affiliation(s)
- István Szokodi
- Department of Pharmacology and Toxicology, Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|