1
|
Liu YB, Wang Q, Song YL, Song XM, Fan YC, Kong L, Zhang JS, Li S, Lv YJ, Li ZY, Dai JY, Qiu ZK. Abnormal phosphorylation / dephosphorylation and Ca 2+ dysfunction in heart failure. Heart Fail Rev 2024; 29:751-768. [PMID: 38498262 DOI: 10.1007/s10741-024-10395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Heart failure (HF) can be caused by a variety of causes characterized by abnormal myocardial systole and diastole. Ca2+ current through the L-type calcium channel (LTCC) on the membrane is the initial trigger signal for a cardiac cycle. Declined systole and diastole in HF are associated with dysfunction of myocardial Ca2+ function. This disorder can be correlated with unbalanced levels of phosphorylation / dephosphorylation of LTCC, endoplasmic reticulum (ER), and myofilament. Kinase and phosphatase activity changes along with HF progress, resulting in phased changes in the degree of phosphorylation / dephosphorylation. It is important to realize the phosphorylation / dephosphorylation differences between a normal and a failing heart. This review focuses on phosphorylation / dephosphorylation changes in the progression of HF and summarizes the effects of phosphorylation / dephosphorylation of LTCC, ER function, and myofilament function in normal conditions and HF based on previous experiments and clinical research. Also, we summarize current therapeutic methods based on abnormal phosphorylation / dephosphorylation and clarify potential therapeutic directions.
Collapse
Affiliation(s)
- Yan-Bing Liu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
- Medical College, Qingdao University, Qingdao, China
| | - Qian Wang
- Medical College, Qingdao University, Qingdao, China
| | - Yu-Ling Song
- Department of Pediatrics, Huantai County Hospital of Traditional Chinese Medicine, Zibo, China
| | | | - Yu-Chen Fan
- Medical College, Qingdao University, Qingdao, China
| | - Lin Kong
- Medical College, Qingdao University, Qingdao, China
| | | | - Sheng Li
- Medical College, Qingdao University, Qingdao, China
| | - Yi-Ju Lv
- Medical College, Qingdao University, Qingdao, China
| | - Ze-Yang Li
- Medical College, Qingdao University, Qingdao, China
| | - Jing-Yu Dai
- Department of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| | - Zhen-Kang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
2
|
Qin X, Lei C, Yan L, Sun H, Liu X, Guo Z, Sun W, Guo X, Fang Q. Proteomic and Metabolomic Analyses of Right Ventricular Failure due to Pulmonary Arterial Hypertension. Front Mol Biosci 2022; 9:834179. [PMID: 35865003 PMCID: PMC9294162 DOI: 10.3389/fmolb.2022.834179] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/20/2022] [Indexed: 11/23/2022] Open
Abstract
Right ventricular failure (RVF) is the independent and strongest predictor of mortality in pulmonary arterial hypertension (PAH), but, at present, there are no preventive and therapeutic strategies directly targeting the failing right ventricle (RV). The underlying mechanism of RV hypertrophy (RVH) and dysfunction needs to be explored in depth. In this study, we used myocardial proteomics combined with metabolomics to elucidate potential pathophysiological changes of RV remodeling in a monocrotaline (MCT)-induced PAH rat model. The proteins and metabolites extracted from the RV myocardium were identified using label-free liquid chromatography–tandem mass spectrometry (LC-MS/MS). The bioinformatic analysis indicated that elevated intracellular Ca2+ concentrations and inflammation may contribute to myocardial proliferation and contraction, which may be beneficial for maintaining the compensated state of the RV. In the RVF stage, ferroptosis, mitochondrial metabolic shift, and insulin resistance are significantly involved. Dysregulated iron homeostasis, glutathione metabolism, and lipid peroxidation related to ferroptosis may contribute to RV decompensation. In conclusion, we depicted a proteomic and metabolomic profile of the RV myocardium during the progression of MCT-induced PAH, and also provided the insights for potential therapeutic targets facilitating the retardation or reversal of RV dysfunction in PAH.
Collapse
Affiliation(s)
- Xiaohan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chuxiang Lei
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Haidan Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoyan Liu
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhengguang Guo
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Xiaoxiao Guo, ; Quan Fang,
| | - Quan Fang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Xiaoxiao Guo, ; Quan Fang,
| |
Collapse
|
3
|
Wang XR, Du HB, Wang HH, Zhang LM, Si YH, Zhang H, Zhao ZG. Mesenteric Lymph Drainage Improves Cardiac Papillary Contractility and Calcium Sensitivity in Rats with Hemorrhagic Shock. J Surg Res 2021; 266:245-253. [PMID: 34034059 DOI: 10.1016/j.jss.2021.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Myocardial dysfunction is an important adverse factor of hemorrhagic shock that induces refractory hypotension, and post-hemorrhagic shock mesenteric lymph (PHSML) return is involved in this adverse effect. This study investigated whether mesenteric lymph drainage (MLD) improves PHSML return-induced cardiac contractile dysfunction via the restoration of cardiomyocyte calcium sensitivity. MATERIALS AND METHODS A hemorrhage shock model was established by using a controlled hemorrhage through the femoral artery that maintained a mean arterial pressure of 40 ± 2 mmHg for 3 h. MLD and mesenteric lymph duct ligation (MLDL) were performed from 1 to 3 h during hypotension. The papillary muscles of the heart were collected for measurement of calmodulin expression and for determining contractile responses to either isoprenaline or calcium. RESULTS The results showed that either MLD or MLDL reversed the hemorrhagic shock-induced downregulation of calmodulin expression, a marker protein of cardiomyocyte calcium sensitization, in papillary muscles. MLD also improved the decreased contractile response and ±df/dt of the papillary muscle strip to gradient isoprenaline or calcium caused by hemorrhagic shock. CONCLUSION These findings indicate that increased cardiac contractibility may be associated with the restoration of calcium sensitivity produced by PHSML drainage.
Collapse
Affiliation(s)
- Xiao-Rong Wang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China
| | - Hui-Bo Du
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China
| | - Huai-Huai Wang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China; The Second Affiliated Hospital, Hebei North University, Zhangjiakou, China
| | - Li-Min Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China
| | - Yong-Hua Si
- Department of Pediatrics, Cangzhou City People's Hospital, Cangzhou, China
| | - Hong Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China; Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China.
| |
Collapse
|
4
|
Kaykı-Mutlu G, Papazisi O, Palmen M, Danser AHJ, Michel MC, Arioglu-Inan E. Cardiac and Vascular α 1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells 2020; 9:E2412. [PMID: 33158106 PMCID: PMC7694190 DOI: 10.3390/cells9112412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
As heart failure (HF) is a devastating health problem worldwide, a better understanding and the development of more effective therapeutic approaches are required. HF is characterized by sympathetic system activation which stimulates α- and β-adrenoceptors (ARs). The exposure of the cardiovascular system to the increased locally released and circulating levels of catecholamines leads to a well-described downregulation and desensitization of β-ARs. However, information on the role of α-AR is limited. We have performed a systematic literature review examining the role of both cardiac and vascular α1-ARs in HF using 5 databases for our search. All three α1-AR subtypes (α1A, α1B and α1D) are expressed in human and animal hearts and blood vessels in a tissue-dependent manner. We summarize the changes observed in HF regarding the density, signaling and responses of α1-ARs. Conflicting findings arise from different studies concerning the influence that HF has on α1-AR expression and function; in contrast to β-ARs there is no consistent evidence for down-regulation or desensitization of cardiac or vascular α1-ARs. Whether α1-ARs are a therapeutic target in HF remains a matter of debate.
Collapse
Affiliation(s)
- Gizem Kaykı-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| | - Olga Papazisi
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - Meindert Palmen
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Martin C. Michel
- Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| |
Collapse
|
5
|
Hartmann S, Ridley AJ, Lutz S. The Function of Rho-Associated Kinases ROCK1 and ROCK2 in the Pathogenesis of Cardiovascular Disease. Front Pharmacol 2015; 6:276. [PMID: 26635606 PMCID: PMC4653301 DOI: 10.3389/fphar.2015.00276] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/03/2015] [Indexed: 01/26/2023] Open
Abstract
Rho-associated kinases ROCK1 and ROCK2 are serine/threonine kinases that are downstream targets of the small GTPases RhoA, RhoB, and RhoC. ROCKs are involved in diverse cellular activities including actin cytoskeleton organization, cell adhesion and motility, proliferation and apoptosis, remodeling of the extracellular matrix and smooth muscle cell contraction. The role of ROCK1 and ROCK2 has long been considered to be similar; however, it is now clear that they do not always have the same functions. Moreover, depending on their subcellular localization, activation, and other environmental factors, ROCK signaling can have different effects on cellular function. With respect to the heart, findings in isoform-specific knockout mice argue for a role of ROCK1 and ROCK2 in the pathogenesis of cardiac fibrosis and cardiac hypertrophy, respectively. Increased ROCK activity could play a pivotal role in processes leading to cardiovascular diseases such as hypertension, pulmonary hypertension, angina pectoris, vasospastic angina, heart failure, and stroke, and thus ROCK activity is a potential new biomarker for heart disease. Pharmacological ROCK inhibition reduces the enhanced ROCK activity in patients, accompanied with a measurable improvement in medical condition. In this review, we focus on recent findings regarding ROCK signaling in the pathogenesis of cardiovascular disease, with a special focus on differences between ROCK1 and ROCK2 function.
Collapse
Affiliation(s)
- Svenja Hartmann
- Institute of Pharmacology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research, Göttingen, Germany
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, UK
| | - Anne J. Ridley
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, UK
| | - Susanne Lutz
- Institute of Pharmacology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research, Göttingen, Germany
| |
Collapse
|
6
|
Zhang X, Zang N, Wei Y, Yin J, Teng R, Seftel A, Disanto ME. Testosterone regulates smooth muscle contractile pathways in the rat prostate: emphasis on PDE5 signaling. Am J Physiol Endocrinol Metab 2012; 302:E243-53. [PMID: 22028410 PMCID: PMC3340899 DOI: 10.1152/ajpendo.00458.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Testosterone (T) plays a permissive role in the development of benign prostatic hyperplasia (BPH), and phosphodiesterase 5 inhibitors (PDE5is) have been found to be effective for BPH and lower urinary tract symptoms (LUTS) in clinical trials. This study investigated the effect of T on smooth muscle (SM) contractile and regulatory signaling pathways, including PDE5 expression and functional activity in prostate in male rats (sham-operated, surgically castrated, and castrated with T supplementation). In vitro organ bath studies, real-time RT-PCR, Western blot analysis, and immunohistochemistry were performed. Castration heavily attenuated contractility, including sensitivity to phenylephrine with SM myosin immunostaining revealing a disrupted SM cell arrangement in the stroma. PDE5 was immunolocalized exclusively in the prostate stroma, and orchiectomy signficantly reduced PDE5 immunopositivity, mRNA, and protein expression, along with nNOS and ROKβ mRNA, whereas it increased eNOS plus α(1a) and α(1b) adrenoreceptor expression in castrated animals. The PDE5i zaprinast significantly increased prostate strip relaxation to the nitric oxide donor sodium nitroprusside (SNP) in control but not castrated rats. But SNP alone was more effective on castrated rats, comparable with sham treated with SNP plus zaprinast. T supplementation prevented or restored all above changes, including SNP and zaprinast in vitro responsiveness. In conclusion, our data show that T positively regulates PDE5 expression and functional activities in prostate, and T ablation not only suppresses prostate size but also reduces prostatic SM contractility, with several potential SM contraction/relaxation pathways implicated. Zaprinast findings strongly suggest a major role for PDE5/cGMP in this signaling cascade. PDE5 inhibition may represent a novel mechanism for treatment of BPH.
Collapse
Affiliation(s)
- Xinhua Zhang
- Cooper University Hospital, Three Cooper Plaza, Camden, NJ 08103, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Nunes KP, Rigsby CS, Webb RC. RhoA/Rho-kinase and vascular diseases: what is the link? Cell Mol Life Sci 2010; 67:3823-36. [PMID: 20668910 PMCID: PMC2996825 DOI: 10.1007/s00018-010-0460-1] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 07/07/2010] [Accepted: 07/08/2010] [Indexed: 12/29/2022]
Abstract
RhoA/Rho-kinase pathway plays an important role in many pathological conditions. RhoA participates in the regulation of smooth muscle tone and activates many downstream kinases. The best characterized are the serine/threonine kinase isoforms (Rho-kinase or ROCK), ROCKα/ROCK2 and ROCKβ/ROCK1. ROCK is necessary for diverse functions such as local blood flow, arterial/pulmonary blood pressure, airway resistance and intestinal peristalsis. ROCK activation permits actin/myosin interactions and smooth muscle cells contraction by maintaining the activity of myosin light-chain kinase, independently of the free cytosolic calcium level. The sensitization of smooth muscle myofilaments to calcium has been implicated in many pathological states, such as hypertension, diabetes, heart attack, stroke, pulmonary hypertension, erectile dysfunction, and cancer. The focus of this review is on the involvement of RhoA/Rho-kinase in diseases. We will briefly describe the ROCK isoforms and the role of RhoA/Rho-kinase in the vasculature, before exploring the most recent findings regarding this pathway and various diseases.
Collapse
Affiliation(s)
- Kenia Pedrosa Nunes
- Department of Physiology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | |
Collapse
|
8
|
Mizutani H, Okamoto R, Moriki N, Konishi K, Taniguchi M, Fujita S, Dohi K, Onishi K, Suzuki N, Satoh S, Makino N, Itoh T, Hartshorne DJ, Ito M. Overexpression of myosin phosphatase reduces Ca(2+) sensitivity of contraction and impairs cardiac function. Circ J 2009; 74:120-8. [PMID: 19966500 DOI: 10.1253/circj.cj-09-0462] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Phosphorylation of the regulatory light chain of myosin (MLC) has roles in cardiac function. In vitro, myosin phosphatase target subunit 2 (MYPT2) is a strongly suspected regulatory subunit of cardiac myosin phosphatase (MP), but there is no in-vivo evidence regarding the functions of MYPT2 in the heart. METHODS AND RESULTS Transgenic mice (Tg) overexpressing MYPT2 were generated using the alpha-MHC promoter. Tg hearts showed an increased expression of MYPT2 and concomitant increase of the endogenous catalytic subunit of type 1 phosphatase (PP1cdelta), resulting in an increase of the MP holoenzyme. The level of phosphorylation of ventricular MLC was reduced. The pCa-tension relationship, using beta-escin permeabilized fibers, revealed decreased Ca(2+) sensitization of contraction in the Tg heart. LV enlargement with associated impairment of function was observed in the Tg heart and ultrastructural examination showed cardiomyocyte degeneration. CONCLUSIONS Overexpression of MYPT2 and the increase in PP1cdelta resulted in an increase of the MP holoenzyme and a decrease in the level of MLC phosphorylation. The latter induced Ca(2+) desensitization of contraction and decreased LV contractility, resulting in LV enlargement. Thus, MYPT2 is truly the regulatory subunit of cardiac MP in-vivo and plays a significant role in modulating cardiac function. (Circ J 2010; 74: 120 - 128).
Collapse
Affiliation(s)
- Hideo Mizutani
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hussain RI, Qvigstad E, Birkeland JAK, Eikemo H, Glende A, Sjaastad I, Skomedal T, Osnes JB, Levy FO, Krobert KA. Activation of muscarinic receptors elicits inotropic responses in ventricular muscle from rats with heart failure through myosin light chain phosphorylation. Br J Pharmacol 2009. [PMID: 19159405 DOI: 10.1111/j.1476-5381.2009.00016.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Muscarinic stimulation increases myofilament Ca(2+) sensitivity with no apparent inotropic response in normal rat myocardium. Increased myofilament Ca(2+) sensitivity is a molecular mechanism promoting increased contractility in failing cardiac tissue. Thus, muscarinic receptor activation could elicit inotropic responses in ventricular myocardium from rats with heart failure, through increasing phosphorylation of myosin light chain (MLC). EXPERIMENTAL APPROACH Contractile force was measured in left ventricular papillary muscles from male Wistar rats, 6 weeks after left coronary artery ligation or sham surgery. Muscles were also frozen, and MLC-2 phosphorylation level was quantified. KEY RESULTS Carbachol (10 micromol.L(-1)) evoked a positive inotropic response only in muscles from rats with heart failure approximating 36% of that elicited by 1 micromol.L(-1) isoproterenol (20 +/- 1.5% and 56 +/- 6.1% above basal respectively). Carbachol-evoked inotropic responses did not correlate with infarction size but did correlate with increased left ventricular end diastolic pressure, heart weight/body weight ratio and lung weight, primary indicators of the severity of heart failure. Only muscarinic receptor antagonists selective for M(2) receptors antagonized carbachol-mediated inotropic effects with the expected potency. Carbachol-evoked inotropic responses and increase in phosphorylated MLC-2 were attenuated by MLC kinase (ML-9) and Rho-kinase inhibition (Y-27632), and inotropic responses were abolished by Pertussis toxin pretreatment. CONCLUSION AND IMPLICATIONS In failing ventricular muscle, muscarinic receptor activation, most likely via M(2) receptors, provides inotropic support by increasing MLC phosphorylation and consequently, myofilament Ca(2+) sensitivity. Enhancement of myofilament Ca(2+) sensitivity, representing a less energy-demanding mechanism of inotropic support may be particularly advantageous in failing hearts.
Collapse
Affiliation(s)
- R I Hussain
- Department of Pharmacology, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Hussain RI, Qvigstad E, Birkeland JAK, Eikemo H, Glende A, Sjaastad I, Skomedal T, Osnes JB, Levy FO, Krobert KA. Activation of muscarinic receptors elicits inotropic responses in ventricular muscle from rats with heart failure through myosin light chain phosphorylation. Br J Pharmacol 2009; 156:575-86. [PMID: 19159405 DOI: 10.1111/j.1750-3639.2009.00016.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Muscarinic stimulation increases myofilament Ca(2+) sensitivity with no apparent inotropic response in normal rat myocardium. Increased myofilament Ca(2+) sensitivity is a molecular mechanism promoting increased contractility in failing cardiac tissue. Thus, muscarinic receptor activation could elicit inotropic responses in ventricular myocardium from rats with heart failure, through increasing phosphorylation of myosin light chain (MLC). EXPERIMENTAL APPROACH Contractile force was measured in left ventricular papillary muscles from male Wistar rats, 6 weeks after left coronary artery ligation or sham surgery. Muscles were also frozen, and MLC-2 phosphorylation level was quantified. KEY RESULTS Carbachol (10 micromol.L(-1)) evoked a positive inotropic response only in muscles from rats with heart failure approximating 36% of that elicited by 1 micromol.L(-1) isoproterenol (20 +/- 1.5% and 56 +/- 6.1% above basal respectively). Carbachol-evoked inotropic responses did not correlate with infarction size but did correlate with increased left ventricular end diastolic pressure, heart weight/body weight ratio and lung weight, primary indicators of the severity of heart failure. Only muscarinic receptor antagonists selective for M(2) receptors antagonized carbachol-mediated inotropic effects with the expected potency. Carbachol-evoked inotropic responses and increase in phosphorylated MLC-2 were attenuated by MLC kinase (ML-9) and Rho-kinase inhibition (Y-27632), and inotropic responses were abolished by Pertussis toxin pretreatment. CONCLUSION AND IMPLICATIONS In failing ventricular muscle, muscarinic receptor activation, most likely via M(2) receptors, provides inotropic support by increasing MLC phosphorylation and consequently, myofilament Ca(2+) sensitivity. Enhancement of myofilament Ca(2+) sensitivity, representing a less energy-demanding mechanism of inotropic support may be particularly advantageous in failing hearts.
Collapse
Affiliation(s)
- R I Hussain
- Department of Pharmacology, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Solaro RJ, Arteaga GM. Heart failure, ischemia/reperfusion injury and cardiac troponin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 592:191-200. [PMID: 17278366 DOI: 10.1007/978-4-431-38453-3_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over the forty years since its discovery, there has been a profound transition in thinking with regard to the role of troponin in the control of cardiac function. This transition involved a change in perception oftroponin as a passive molecular switch responding to membrane controlled fluctuations in cytoplasmic Ca2+ to a perception of troponin as a critical element in signaling cascades that actively engage in control of cardiac function. Evidence demonstrating functionally significant developmental and mutant isoform switches and post-translational modifications of cardiac troponin complex proteins, troponin I (cTnI) and troponin T (cTnT) provided convincing evidence for a more complicated role of troponin in control of cardiac function and dynamics. The physiological role of these modifications of troponin is reviewed in this monograph and has also been reviewed elsewhere (Solaro and Rarick, 1998; Gordon et al., 2000; Solaro et al., 2002a; Kobayashi and Solaro, 2005). Our focus here is on studies related to modifications in troponin that appear important in the processes leading from compensated hypertrophy to heart failure. These studies reveal the potentially significant role of post-translational modifications of troponin in these processes. Another focus is on troponin as a target for inotropic agents. Pharmacological manipulation of troponin by small molecules remains an important avenue of approach for the treatment of acute and chronic heart failure (Kass and Solaro, 2006).
Collapse
Affiliation(s)
- R John Solaro
- Department of Physiology and Biophysics (M/C 901), University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | | |
Collapse
|
12
|
Grimm M, El-Armouche A, Zhang R, Anderson ME, Eschenhagen T. Reduced contractile response to α1-adrenergic stimulation in atria from mice with chronic cardiac calmodulin kinase II inhibition. J Mol Cell Cardiol 2007; 42:643-52. [PMID: 17292391 DOI: 10.1016/j.yjmcc.2006.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Revised: 12/15/2006] [Accepted: 12/20/2006] [Indexed: 10/01/2022]
Abstract
The sustained positive inotropic effect of alpha-adrenoceptor agonists in the heart is associated with a small increase in intracellular Ca(2+) transients together with a larger sensitization of myofilaments to Ca(2+). The multifunctional Ca(2+) and calmodulin-dependent protein kinase II (CaMKII) could contribute to this effect, either by affecting the Ca(2+) release (ryanodine receptor) or by an uptake mechanism (via phospholamban [PLB] and SR Ca(2+) ATPase). Here we examined the role of CaMKII in the positive inotropic effect of the alpha-adrenoceptor agonist phenylephrine in left atria isolated from a genetic mouse model of cardiac CaMKII inhibition (AC3-I). Compared to atria from wild-type (WT) or AC3-C (scrambled peptide), AC3-I atria showed the following abnormalities. PLB phosphorylation at Thr17, a known CaMKII target, was significantly lower ( approximately 20%). Post-rest (30 s, 1 Hz, 37 degrees C) potentiation of force was absent (AC3-C, 190% of pre-rest amplitude). Basal force was approximately 20% lower at 1.8 mM Ca(2+), but normal at high Ca(2+) concentration (>4.5 mM). The maximal positive inotropic effect of phenylephrine, which was more pronounced at low frequencies in WT and AC3-C atria, lost its frequency dependence (1 Hz to 8 Hz). Thus, the effect of phenylephrine was reduced by approximately 50% at 1 Hz, but was normal at 8 Hz. All three groups showed a negative force-frequency relation, and did not differ in the frequency-dependent acceleration of relaxation. Our data indicate a role of CaMKII in post-rest potentiation and the positive inotropic effect of alpha-adrenergic stimulation at low frequencies.
Collapse
Affiliation(s)
- Michael Grimm
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | |
Collapse
|
13
|
Hu E, Lee D. Rho kinase as potential therapeutic target for cardiovascular diseases: opportunities and challenges. Expert Opin Ther Targets 2007; 9:715-36. [PMID: 16083339 DOI: 10.1517/14728222.9.4.715] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Rho kinase (ROCK) belongs to a family of Ser/Thr protein kinases that are activated via interaction with the small GTP-binding protein RhoA. Growing evidence suggests that RhoA and ROCK participate in a variety of important physiological functions in vasculature including smooth muscle contraction, cell proliferation, cell adhesion and migration, and many aspects of inflammatory responses. As these processes mediate the onset and progression of cardiovascular disease, modulation of the Rho/ROCK signalling pathway is a potential strategy for targeting an array of cardiovascular indications. Two widely employed ROCK inhibitors, fasudil and Y-27632, have provided preliminary but compelling evidence supporting the potential cardiovascular benefits of ROCK inhibition in preclinical animal disease models and in the clinic. This review summarises the molecular biology of ROCK and its biological functions in smooth muscle, endothelium and other vascular tissues. In addition, there will be a focus on recent progress demonstrating the benefits of ROCK inhibition in several animal models of cardiovascular diseases. Finally, recent progress in the identification of novel ROCK inhibitors and challenges associated with their development for clinical use will be discussed.
Collapse
Affiliation(s)
- Erding Hu
- Center of Excellence for Cardiovascular and Urogenital Drug Discovery, Department of Vascular Biology, GlaxoSmithKline Pharmaceuticals, 709 Swedeland Road, King of Prussia, PA 19406, USA.
| | | |
Collapse
|
14
|
Machackova J, Barta J, Dhalla NS. Myofibrillar remodeling in cardiac hypertrophy, heart failure and cardiomyopathies. Can J Cardiol 2006; 22:953-68. [PMID: 16971981 PMCID: PMC2570240 DOI: 10.1016/s0828-282x(06)70315-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Accepted: 06/20/2006] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND A wide variety of pathological conditions have been shown to result in cardiac remodelling and myocardial dysfunction. However, the mechanisms of transition from adaptive to maladaptive alterations, as well as those for changes in cardiac performance leading to heart failure, are poorly understood. OBSERVATIONS Extensive studies have revealed a broad spectrum of progressive changes in subcellular structures and function, as well as in signal transduction and metabolism in the heart, among different cardiovascular disorders. The present review is focused on identifying the alterations in molecular and biochemical structure of myofibrils (myofibrillar remodelling) in hypertrophied and failing myocardium in different types of heart diseases. Numerous changes at the level of gene expression for both contractile and regulatory proteins have already been reported in failing hearts and heart diseases; these changes are potential precursors for heart failure such as cardiac hypertrophy and cardiomyopathies. Myofibrillar remodelling, as a consequence of proteolysis, oxidation, and phosphorylation of some functional groups in both contractile and regulatory proteins in hearts failing due to different etiologies, has also been described. CONCLUSIONS Although myofibrillar remodelling appears to be associated with cardiac dysfunction, alterations in both contractile and regulatory proteins are dependent on the type and stage of heart disease.
Collapse
Affiliation(s)
- Jarmila Machackova
- Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba
| | - Judit Barta
- Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba
| |
Collapse
|
15
|
Hirano S, Kusakari Y, O-Uchi J, Morimoto S, Kawai M, Hongo K, Kurihara S. Intracellular mechanism of the negative inotropic effect induced by alpha1-adrenoceptor stimulation in mouse myocardium. J Physiol Sci 2006; 56:297-304. [PMID: 16884559 DOI: 10.2170/physiolsci.rp007306] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2006] [Accepted: 08/02/2006] [Indexed: 11/05/2022]
Abstract
Alpha(1)-adrenoceptor stimulation (alpha(1)ARS) shows a positive inotropic effect in most mammalian myocardium. In mouse myocardium, however, alpha(1)ARS showed the negative inotropic effect, of which intracellular mechanisms are not fully clarified. The purpose of this study is to investigate the intracellular mechanism of the negative inotropic effect by alpha(1)ARS in C57BL/6 mouse myocardium. We used isolated ventricular papillary muscles of C57BL/6 strain mouse which is widely used for genetic manipulation. We simultaneously measured isometric tension and intracellular Ca(2+) concentration ([Ca(2+)](i)) using the aequorin method. In twitch contraction, phenylephrine concentration-dependently (1-100 microM) decreased tension without significant changes in the Ca(2+) transient, and these effects were completely blocked by prazosin (3 microM) or calphostin C (a PKC inhibitor, 1 microM). Phorbol 12-myristate 13-acetate (PMA) (a PKC activator, 1 microM) decreased tension as observed in phenylephrine. After PMA application, the negative inotropic effect of phenylephrine disappeared. To estimate the Ca(2+) sensitivity, tetanic contraction was produced, and the relation between [Ca(2+)](i) and tension at a steady state was measured. Phenylephrine (10 microM) decreased the Ca(2+) sensitivity, and PMA showed a similar Ca(2+) desensitizing effect. These results suggest that the negative inotropic effect of phenylephrine in mouse myocardium can be explained by the decrease in the Ca(2+) sensitivity through the activation of PKC. The present result indicates that the effect of alpha(1)ARS differs among species and strains of experiment animals. Thus, we should be careful about using the results of mouse myocardium to understand the functions of the human heart.
Collapse
Affiliation(s)
- Shuta Hirano
- Department of Physiology II, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Okamoto R, Kato T, Mizoguchi A, Takahashi N, Nakakuki T, Mizutani H, Isaka N, Imanaka-Yoshida K, Kaibuchi K, Lu Z, Mabuchi K, Tao T, Hartshorne DJ, Nakano T, Ito M. Characterization and function of MYPT2, a target subunit of myosin phosphatase in heart. Cell Signal 2006; 18:1408-16. [PMID: 16431080 DOI: 10.1016/j.cellsig.2005.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Revised: 11/09/2005] [Accepted: 11/09/2005] [Indexed: 11/19/2022]
Abstract
Characterization of cardiac MYPT2 (an isoform of the smooth muscle phosphatase [MP] target subunit, MYPT1) is described. Several features of MYPT2 and MYPT1 were similar, including: a specific interaction with the catalytic subunit of type 1 phosphatase, delta isoform (PP1cdelta); interaction of MYPT2 with the small heart-specific MP subunit; interaction of the C-terminal region of MYPT2 with the active form of RhoA; phosphorylation by Rho-kinase at an inhibitory site, Thr646 and thiophosphorylation at Thr646 inhibited activity of the MYPT2-PP1cdelta complex. MYPT2 activated PP1cdelta activity, using light chains from smooth and cardiac muscle, by reducing K(m) and increasing k(cat). The extent of activation (k(cat)) was greater than for MYPT1 and could reflect distinct N-terminal sequences in the two MYPT isoforms. Adenovirus-mediated gene transfer of MYPT2 and PP1cdelta reduced the phosphorylation level of cardiac light chains following stimulation with A23187. Overexpression of MYPT2 and PP1cdelta blocked the angiotensin II-induced sarcomere organization in cultured cardiomyocytes. Electron microscopy indicated locations of MYPTs, at, or close to, the Z-line, the A band and mitochondria. Similarity of the two MYPT isoforms suggests common enzymatic mechanisms and regulation. Cardiac myosin is a substrate for the MYPT2 holoenzyme, but the Z-line location raises the possibility of other substrates.
Collapse
Affiliation(s)
- Ryuji Okamoto
- Department of Cardiology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Machackova J, Barta J, Dhalla NS. Molecular defects in cardiac myofibrillar proteins due to thyroid hormone imbalance and diabetesThis paper is a part of a series in the Journal's "Made in Canada" section. The paper has undergone peer review. Can J Physiol Pharmacol 2005; 83:1071-91. [PMID: 16462907 DOI: 10.1139/y05-121] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The heart very often becomes a victim of endocrine abnormalities such as thyroid hormone imbalance and insulin deficiency, which are manifested in a broad spectrum of cardiac dysfunction from mildly compromised function to severe heart failure. These functional changes in the heart are largely independent of alterations in the coronary arteries and instead reside at the level of cardiomyocytes. The status of cardiac function reflects the net of underlying subcellular modifications induced by an increase or decrease in thyroid hormone and insulin plasma levels. Changes in the contractile and regulatory proteins constitute molecular and structural alterations in myofibrillar assembly, called myofibrillar remodeling. These alterations may be adaptive or maladaptive with respect to the functional and metabolic demands on the heart as a consequence of the altered endocrine status in the body. There is a substantial body of information to indicate alterations in myofibrillar proteins including actin, myosin, tropomyosin, troponin, titin, desmin, and myosin-binding protein C in conditions such as hyperthyroidism, hypothyroidism, and diabetes. The present article is focussed on discussion how myofibrillar proteins are altered in response to thyroid hormone imbalance and lack of insulin or its responsiveness, and how their structural and functional changes explain the contractile defects in the heart.
Collapse
Affiliation(s)
- Jarmila Machackova
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Tache Avenue, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | | | | |
Collapse
|
18
|
Rajashree R, Blunt BC, Hofmann PA. Modulation of myosin phosphatase targeting subunit and protein phosphatase 1 in the heart. Am J Physiol Heart Circ Physiol 2005; 289:H1736-43. [PMID: 15908465 DOI: 10.1152/ajpheart.00318.2004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myosin light chain 2 (LC2) phosphorylation is of both physiological and pathological importance to myocardial function. The phosphatase that directly dephosphorylates LC2 is a type 1 protein phosphatase (PP1) that contains a catalytic subunit that complexes with a myosin-binding phosphatase targeting subunit (MYPT). The goal of the present study was to examine the role of MYPT in the regulation of PP1 in ventricular myocytes. In the first part of the study, regional distribution of MYPT expression and phosphorylation were determined in unstimulated hearts. The pattern of MYPT phosphorylation was inversely related to the LC2 phosphorylation spatial gradient as described by Epstein and colleagues (Davis JS, Hassanzadeh S, Winitsky S, Lin H, Satorius C, Vemuri R, Aletras AH, Wen H, and Epstein ND. Cell 107: 631-641, 2001). In the second part of the study, adult rat isolated ventricular myocytes were exposed to an alpha-adrenergic receptor agonist, and properties of MYPT, PP1, and LC2 were studied. We found MYPT associates with cardiac myofilaments, and this association increases upon alpha-adrenergic receptor stimulation. Activation of alpha-adrenergic receptors also led to a decrease in the PP1-myofilament association. Furthermore, alpha-adrenergic receptor stimulation results in phosphorylation of MYPT and LC2 and an increase in myocyte Ca(2+) sensitivity of tension that all depend on Rho kinase activation. These data support the hypothesis that alpha-adrenergic receptor activation works through Rho kinase to phosphorylate MYPT, and phosphorylated MYPT dissociates from PP1 so that PP1 is no longer physically associated with LC2. Hence, we propose a pathway for the dynamic modulation of LC2 phosphorylation through receptor-dependent phosphorylation of MYPT, and a spatial gradient of LC2 phosphorylation under basal conditions that occurs due to varied levels of phosphorylation of MYPT in ventricles.
Collapse
Affiliation(s)
- Ravi Rajashree
- Department of Physiology, University of Tennessee School of Medicine, 894 Union Ave., Memphis, Tennessee 38163, USA
| | | | | |
Collapse
|
19
|
Chu L, Norota I, Endoh M. Differential inhibition by the Rho kinase inhibitor Y-27632 of the increases in contractility and Ca2+ transients induced by endothelin-1 in rabbit ventricular myocytes. Naunyn Schmiedebergs Arch Pharmacol 2005; 371:185-94. [PMID: 15900512 DOI: 10.1007/s00210-005-1032-z] [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] [Received: 11/02/2004] [Accepted: 01/28/2005] [Indexed: 01/08/2023]
Abstract
The role of Rho kinase activation in the regulation of cardiac contractility and Ca(2+) signaling remains unclear, whereas its role in smooth muscle regulation has been well documented. To study the potential role of Rho kinase in the regulation of cardiac contractility and Ca(2+) transients induced by endothelin-1 (ET-1) and isoproterenol, we used the Rho kinase inhibitor Y-27632 in rabbit ventricular myocardium and myocytes loaded with indo-1/AM. Y-27632 (3-30 microM) inhibited significantly the baseline contractility and Ca(2+) transients. Furthermore, Y-27632 suppressed the increase in contractility and Ca(2+) transients induced by ET-1 in a concentration-dependent manner, when it was used in a concentration at which it did not affect the effects of isoproterenol via beta-adrenoceptors. In the presence of Y-27632, ET-1 increased cell shortening in the absence of an increase in Ca(2+) transients. This is an indication that the increase in myofilament Ca(2+) sensitivity induced by ET-1 is less susceptible to the inhibitory action of Y-27632. These findings imply that the Rho kinase activation may partially contribute to the ET-1-induced regulation of contractility, primarily due to an ET-1-induced increase in Ca(2+) transients in rabbit ventricular myocardium.
Collapse
Affiliation(s)
- Li Chu
- Department of Cardiovascular Pharmacology, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | | | | |
Collapse
|
20
|
Vahebi S, Kobayashi T, Warren CM, de Tombe PP, Solaro RJ. Functional effects of rho-kinase-dependent phosphorylation of specific sites on cardiac troponin. Circ Res 2005; 96:740-7. [PMID: 15774859 DOI: 10.1161/01.res.0000162457.56568.7d] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We tested the hypothesis that activation of Rho-A-dependent kinase (ROCK-II) alters cardiac myofilament response to Ca2+ by mechanisms involving phosphorylation of thin filament proteins. We determined effects of a constitutively active form of ROCK-II on ATPase activity and tension development in detergent-extracted (skinned) fiber bundles isolated from mouse left ventricular papillary muscles. ROCK-II induced a depression in maximum ATPase rate and tension, which was associated with phosphorylation of troponin T (TnT), troponin I (TnI), and myosin-binding protein C (C-protein). This effect of ROCK-II was retained in fiber bundles isolated from transgenic (TG) mice in which phosphorylation sites (S14, S15, and S19) of myosin light chain 2 were mutated to alanine. Moreover, exchange of ROCK-II-phosphorylated Tn complex with the native Tn complex in the fiber bundles resulted in inhibition of maximal Ca2+ activation of tension and ATPase activity. Mass spectrometric analysis demonstrated that ROCK-II phosphorylated cardiac TnI (cTnI) at S23, S24, and T144 and cardiac TnT (cTnT) at S278 and T287. An important role for these cTnT sites is indicated by results demonstrating that ROCK-II induced a depression in tension and ATPase activity in skinned fiber bundles from a TG model in which cTnI is replaced by slow skeletal TnI, which lacks S23 and S24 and in which T144 is replaced by proline. Our data provide the first evidence that ROCK-II phosphorylation of the Tn complex, most likely at cTnT, has an important role in functional effects of signaling through the Rho-A pathway.
Collapse
Affiliation(s)
- Susan Vahebi
- University of Illinois at Chicago Department of Physiology and Biophysics and Center for Cardiovascular Research College of Medicine, Chicago, Ill 60612-7342, USA
| | | | | | | | | |
Collapse
|
21
|
Torsoni AS, Fonseca PM, Crosara-Alberto DP, Franchini KG. Early activation of p160ROCK by pressure overload in rat heart. Am J Physiol Cell Physiol 2003; 284:C1411-9. [PMID: 12570982 DOI: 10.1152/ajpcell.00098.2002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We investigated the effects of acute pressure overload on activation of p160(ROCK) in rat myocardium. Constriction of transverse aorta, controlled to increase peak systolic pressure of ascending aorta by approximately 40 mmHg, induced a rapid association of RhoA with Dbl-3 and p160(ROCK). The binding of p160(ROCK) to RhoA was rapidly increased, peaking at 30 min (approximately 3.5-fold), but reduced to lower levels (approximately 1.9-fold) by 60 min of pressure overload. The activity of immunoprecipitated p160(ROCK) toward myosin light chain increased approximately 2.5-fold within 10 min but decreased to lower levels (approximately 1.6-fold) after 60 min of pressure overload. Confocal microscopic analysis indicated that pressure overload induced the formation of aggregates of p160(ROCK) and RhoA along the longitudinal axis of cardiac myocytes. Immunoelectron microscopic analysis showed that pressure overload induced the association of p160(ROCK) and RhoA to Z-line, T-tubule, and subsarcolemmal areas. The rapid activation of p160(ROCK) by pressure overload and its aggregation in subcellular structures involved in transmission of mechanical force suggest a role for this enzyme in the mechanobiochemical transduction in the myocardium.
Collapse
Affiliation(s)
- Adriana S Torsoni
- Department of Internal Medicine, School of Medicine, State University of Campinas, 13081-970 Campinas, SP, Brazil
| | | | | | | |
Collapse
|
22
|
Activation of Adenosine A1 and A3 Receptors Protects Mitochondria during Hypoxia in Cardiomyocytes by Distinct Mechanisms. ACTA ACUST UNITED AC 2003. [DOI: 10.1007/978-1-4615-0355-2_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
23
|
Formigli L, Meacci E, Vassalli M, Nosi D, Quercioli F, Tiribilli B, Tani A, Squecco R, Francini F, Bruni P, Zecchi Orlandini S. Sphingosine 1-phosphate induces cell contraction via calcium-independent/Rho-dependent pathways in undifferentiated skeletal muscle cells. J Cell Physiol 2003; 198:1-11. [PMID: 14584038 DOI: 10.1002/jcp.10366] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have previously shown that sphingosine 1-phosphate (S1P) can induce intracellular Ca(2+) mobilization and cell contraction in C2C12 myoblasts and that the two phenomena are temporally unrelated. Although Ca(2+)-independent mechanisms of cell contraction have been the focus of numerous studies on Ca(2+) sensitization of smooth muscle, comparatively less studies have focused on the role that these mechanisms play in the regulation of skeletal muscle contractility. Phosphorylation and activation of myosin by Rho-dependent kinase mediate most of Ca(2+)-independent contractile responses. In the present study, we examined the potential role of Rho/Rho-kinase cascade activation in S1P-induced C2C12 cell contraction. First, we showed that depletion of Ca(2+), by pre-treatment with BAPTA, did not affect S1P-induced myoblastic contractility, whereas it abolished S1P-induced Ca(2+) transients. These results correlated with the absence of troponin C and with the immature cytoskeletal organization of these cells. Experimental evidence demonstrating the involvement of Rho pathway in S1P-stimulated myoblast contraction included: the activation/translocation of RhoA to the membrane in response to agonist-stimulation in cells depleted of Ca(2+) and the inhibition of dynamic changes of the actin cytoskeleton in cells where Rho functions had been inhibited either by overexpression of RhoGDI, a physiological inhibitor of GDP dissociation from Rho proteins, or by pretreatment with Y-27632, a specific Rho kinase inhibitor. Contribution of protein kinase C in this cytoskeletal rearrangement was also evaluated. However, the pretreatment with Gö6976 or rottlerin, specific inhibitors of PKC alpha and PKC delta, respectively, failed to inhibit the agonist-induced myoblastic contraction. Single particle tracking of G-actin fluorescent probe was performed to statistically evaluate actin cytoskeletal dynamics in response to S1P. Stimulation with S1P was also able to increase the phosphorylation level of myosin light chain II. In conclusion, our results strongly suggest that Ca(2+)-independent/Rho-Rho kinase-dependent pathways may exert an important role in S1P-induced myoblastic cell contraction.
Collapse
Affiliation(s)
- L Formigli
- Department of Anatomy, Histology, Forensic Medicine, University of Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
|