1
|
Polyunsaturated ω3 fatty acids prevent the cardiac hypertrophy in hypertensive rats. Biochim Biophys Acta Gen Subj 2023; 1867:130278. [PMID: 36410610 DOI: 10.1016/j.bbagen.2022.130278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/10/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022]
Abstract
It has been demonstrated that supplementation with the two main omega 3 polyunsaturated fatty acids (ω3 FAs), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA), leads to modifications in the cardiac physiology. ω3 FAs can affect the membrane's lipid composition, as well as proteins' location and/or function. The Na+/H+ exchanger (NHE1) is an integral membrane protein involved in the maintenance of intracellular pH and its hyperactivity has been associated with the development of various cardiovascular diseases such as cardiac hypertrophy. Our aim was to determine the effect of ω3 FAs on systolic blood pressure (SBP), lipid profiles, NHE1 activity, and cardiac function in spontaneously hypertensive rats (SHR) using Wistar rats (W) as normotensive control. After weaning, the rats received orally ω3 FAs (200 mg/kg body mass/day/ 4 months). We measured SBP, lipid profiles, and different echocardiography parameters, which were used to calculate cardiac hypertrophy index, systolic function, and ventricular geometry. The rats were sacrificed, and ventricular cardiomyocytes were obtained to measure NHE1 activity. While the treatment with ω3 FAs did not affect the SBP, lipid analysis of plasma revealed a significant decrease in omega-6/omega-3 ratio, correlated with a significant reduction in left ventricular mass index in SHR. The NHE1 activity was significantly higher in SHR compared with W. While in W the NHE1 activity was similar in both groups, a significant decrease in NHE1 activity was detected in SHRs supplemented with ω3 FAs, reaching values comparable with W. Altogether, these findings revealed that diet supplementation with ω3 FAs since early age prevents the development of cardiac hypertrophy in SHR, perhaps by decreasing NHE1 activity, without altering hemodynamic overload.
Collapse
|
2
|
Karmazyn M, Pierce GN, Fliegel L. The Remaining Conundrum of the Role of the Na +/H + Exchanger Isoform 1 (NHE1) in Cardiac Physiology and Pathology: Can It Be Rectified? Rev Cardiovasc Med 2022; 23:284. [PMID: 39076631 PMCID: PMC11266974 DOI: 10.31083/j.rcm2308284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 07/31/2024] Open
Abstract
The mammalian Na + /H + exchanger (NHE) is a family of ubiquitous membrane proteins present in humans. Isoform one (NHE1) is present on the plasma membrane and regulates intracellular pH by removal of one intracellular proton in exchange for one extracellular sodium thus functioning as an electroneutral process. Human NHE1 has a 500 amino acid membrane domain plus a C-terminal 315 amino acid, regulatory cytosolic tail. It is regulated through a cytosolic regulatory C-terminal tail which is subject to phosphorylation and is modulated by proteins and lipids. Substantial evidence has implicated NHE1 activity in both myocardial ischemia and reperfusion damage and myocardial remodeling resulting in heart failure. Experimental data show excellent cardioprotection with NHE1 inhibitors although results from clinical results have been mixed. In cardiac surgery patients receiving the NHE1 inhibitor cariporide, subgroups showed beneficial effects of treatment. However, in one trial this was associated with a significantly increased incidence of ischemic strokes. This likely reflected both inappropriate dosing regimens as well as overly high drug doses. We suggest that further progress towards NHE1 inhibition as a treatment for cardiovascular disease is warranted through the development of novel compounds to inhibit NHE1 that are structurally different than those previously used in compromised clinical trials. Some novel pyrazinoyl guanidine inhibitors of NHE1 are already in development and the recent elucidation of the three-dimensional structure of the NHE1 protein and identity of the inhibitor binding site may facilitate development. An alternative approach may also be to control the endogenous regulation of activity of NHE1, which is activated in disease.
Collapse
Affiliation(s)
- Morris Karmazyn
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Grant N. Pierce
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, and Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
3
|
Zavala MR, Díaz RG, Villa-Abrille MC, Pérez NG. Thioredoxin 1 (TRX1) Overexpression Cancels the Slow Force Response (SFR) Development. Front Cardiovasc Med 2021; 8:622583. [PMID: 33718450 PMCID: PMC7952880 DOI: 10.3389/fcvm.2021.622583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/18/2021] [Indexed: 11/28/2022] Open
Abstract
The stretch of cardiac muscle increases developed force in two phases. The first phase occurs immediately after stretch and is the expression of the Frank–Starling mechanism, while the second one or slow force response (SFR) occurs gradually and is due to an increase in the calcium transient amplitude. An important step in the chain of events leading to the SFR generation is the increased production of reactive oxygen species (ROS) leading to redox sensitive ERK1/2, p90RSK, and NHE1 phosphorylation/activation. Conversely, suppression of ROS production blunts the SFR. The purpose of this study was to explore whether overexpression of the ubiquitously expressed antioxidant molecule thioredoxin-1 (TRX1) affects the SFR development and NHE1 phosphorylation. We did not detect any change in basal phopho-ERK1/2, phopho-p90RSK, and NHE1 expression in mice with TRX1 overexpression compared to wild type (WT). Isolated papillary muscles from WT or TRX1-overexpressing mice were stretched from 92 to 98% of its maximal length. A prominent SFR was observed in WT mice that was completely canceled in TRX1 animals. Interestingly, myocardial stretch induced a significant increase in NHE1 phosphorylation in WT mice that was not detected in TRX1-overexpressing mice. These novel results suggest that magnification of cardiac antioxidant defense power by overexpression of TRX1 precludes NHE1 phosphorylation/activation after stretch, consequently blunting the SFR development.
Collapse
Affiliation(s)
- Maite R Zavala
- Fellow From Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Romina G Díaz
- Established Investigators of CONICET, Buenos Aires, Argentina
| | - María C Villa-Abrille
- Established Investigators of CONICET, Buenos Aires, Argentina.,Full Professors of Physiology, Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Néstor G Pérez
- Established Investigators of CONICET, Buenos Aires, Argentina.,Full Professors of Physiology, Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, Buenos Aires, Argentina
| |
Collapse
|
4
|
Ennis IL, Pérez NG. Cardiac Mineralocorticoid Receptor and the Na +/H + Exchanger: Spilling the Beans. Front Cardiovasc Med 2021; 7:614279. [PMID: 33553262 PMCID: PMC7854694 DOI: 10.3389/fcvm.2020.614279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023] Open
Abstract
Current evidence reveals that cardiac mineralocorticoid receptor (MR) activation following myocardial stretch plays an important physiological role in adapting developed force to sudden changes in hemodynamic conditions. Its underlying mechanism involves a previously unknown nongenomic effect of the MR that triggers redox-mediated Na+/H+ exchanger (NHE1) activation, intracellular Na+ accumulation, and a consequent increase in Ca2+ transient amplitude through reverse Na+/Ca2+ exchange. However, clinical evidence assigns a detrimental role to MR activation in the pathogenesis of severe cardiac diseases such as congestive heart failure. This mini review is meant to present and briefly discuss some recent discoveries about locally triggered cardiac MR signals with the objective of shedding some light on its physiological but potentially pathological consequences in the heart.
Collapse
Affiliation(s)
- Irene Lucía Ennis
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas de la Plata, Universidad Nacional de La Plata, La Plata, Argentina
| | - Néstor Gustavo Pérez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas de la Plata, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
5
|
Brea MS, Díaz RG, Escudero DS, Zavala MR, Portiansky EL, Villa-Abrille MC, Caldiz CI, Pérez NG, Morgan PE. Silencing of epidermal growth factor receptor reduces Na+/H+ exchanger 1 activity and hypertensive cardiac hypertrophy. Biochem Pharmacol 2019; 170:113667. [DOI: 10.1016/j.bcp.2019.113667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
|
6
|
Dowrick JM, Tran K, Loiselle DS, Nielsen PMF, Taberner AJ, Han J, Ward M. The slow force response to stretch: Controversy and contradictions. Acta Physiol (Oxf) 2019; 226:e13250. [PMID: 30614655 DOI: 10.1111/apha.13250] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/20/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
When exposed to an abrupt stretch, cardiac muscle exhibits biphasic active force enhancement. The initial, instantaneous, force enhancement is well explained by the Frank-Starling mechanism. However, the cellular mechanisms associated with the second, slower phase remain contentious. This review explores hypotheses regarding this "slow force response" with the intention of clarifying some apparent contradictions in the literature. The review is partitioned into three sections. The first section considers pathways that modify the intracellular calcium handling to address the role of the sarcoplasmic reticulum in the mechanism underlying the slow force response. The second section focuses on extracellular calcium fluxes and explores the identity and contribution of the stretch-activated, non-specific, cation channels as well as signalling cascades associated with G-protein coupled receptors. The final section introduces promising candidates for the mechanosensor(s) responsible for detecting the stretch perturbation.
Collapse
Affiliation(s)
- Jarrah M. Dowrick
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Kenneth Tran
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Denis S. Loiselle
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Physiology University of Auckland Auckland New Zealand
| | - Poul M. F. Nielsen
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - Andrew J. Taberner
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - June‐Chiew Han
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Marie‐Louise Ward
- Department of Physiology University of Auckland Auckland New Zealand
| |
Collapse
|
7
|
Yamaguchi Y, Iribe G, Kaneko T, Takahashi K, Numaga-Tomita T, Nishida M, Birnbaumer L, Naruse K. TRPC3 participates in angiotensin II type 1 receptor-dependent stress-induced slow increase in intracellular Ca 2+ concentration in mouse cardiomyocytes. J Physiol Sci 2018; 68:153-164. [PMID: 28105583 PMCID: PMC10718017 DOI: 10.1007/s12576-016-0519-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/26/2016] [Indexed: 11/26/2022]
Abstract
When a cardiac muscle is held in a stretched position, its [Ca2+] transient increases slowly over several minutes in a process known as stress-induced slow increase in intracellular Ca2+ concentration ([Ca2+]i) (SSC). Transient receptor potential canonical (TRPC) 3 forms a non-selective cation channel regulated by the angiotensin II type 1 receptor (AT1R). In this study, we investigated the role of TRPC3 in the SSC. Isolated mouse ventricular myocytes were electrically stimulated and subjected to sustained stretch. An AT1R blocker, a phospholipase C inhibitor, and a TRPC3 inhibitor suppressed the SSC. These inhibitors also abolished the observed SSC-like slow increase in [Ca2+]i induced by angiotensin II, instead of stretch. Furthermore, the SSC was not observed in TRPC3 knockout mice. Simulation and immunohistochemical studies suggest that sarcolemmal TRPC3 is responsible for the SSC. These results indicate that sarcolemmal TRPC3, regulated by AT1R, causes the SSC.
Collapse
Affiliation(s)
- Yohei Yamaguchi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Gentaro Iribe
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan.
| | - Toshiyuki Kaneko
- Department of Physiology, Asahikawa Medical University, Asahikawa, Hokkaido, 078-8510, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Takuro Numaga-Tomita
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Science, Research Triangle Park, NC, 27709, USA
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| |
Collapse
|
8
|
Arginine-vasopressin therapy in hypotensive neonates and infants after cardiac surgery: response is unrelated to baseline ventricular function. Cardiol Young 2017; 27:1031-1040. [PMID: 27964765 DOI: 10.1017/s104795111600189x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We hypothesised that infants with ventricular dysfunction after cardiac surgery have impaired haemodynamic response to arginine-vasopressin therapy. We retrospectively reviewed the medical records of neonates and infants treated with arginine-vasopressin within 48 hours of corrective or palliative cardiac surgery who underwent echocardiographic assessment of ventricular function before initiation of therapy. Patients were classified as "responders" if their systolic blood pressure increased by ⩾10% without increase in catecholamine score or if it was maintained with decreased catecholamine score. Response was assessed 1 hour after maximum upward titration of arginine-vasopressin. A total of 36 children (15 neonates) were reviewed (17 male). The median (interquartile) age was 10.4 weeks (1.1-26.9), and the median weight was 4.3 kg (3.2-5.8). Diagnoses included single ventricle (eight), arch abnormalities (five), atrioventricular septal defect (four), double-outlet right ventricle (three), tetralogy of Fallot (three), and others (13). In all, 12 patients (33%) had ventricular dysfunction. Only 15 (42%) responded favourably according to our definition 1 hour after the "target" arginine-vasopressin dose was achieved. Ventricular dysfunction was not associated with poor response. The overall mortality was 25%, but mortality in patients with ventricular dysfunction was 42%. Favourable response was associated with shorter ICU stay (9.5 days versus 19.5 days, p=0.01). We conclude that arginine-vasopressin fails to increase blood pressure in ~50% of hypotensive children after cardiac surgery. The response rate does not increase with duration of therapy. Ventricular function does not predict haemodynamic response. The mortality in this group is very high. Prospective comparison of vasopressin with other vasoactive agents and/or inotropes is warranted.
Collapse
|
9
|
Transactivation of the epidermal growth factor receptor in responses to myocardial stress and cardioprotection. Int J Biochem Cell Biol 2017; 83:97-110. [PMID: 28049018 DOI: 10.1016/j.biocel.2016.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/25/2016] [Accepted: 12/26/2016] [Indexed: 12/20/2022]
|
10
|
β-Arrestin mediates the Frank-Starling mechanism of cardiac contractility. Proc Natl Acad Sci U S A 2016; 113:14426-14431. [PMID: 27911784 DOI: 10.1073/pnas.1609308113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Frank-Starling law of the heart is a physiological phenomenon that describes an intrinsic property of heart muscle in which increased cardiac filling leads to enhanced cardiac contractility. Identified more than a century ago, the Frank-Starling relationship is currently known to involve length-dependent enhancement of cardiac myofilament Ca2+ sensitivity. However, the upstream molecular events that link cellular stretch to the length-dependent myofilament Ca2+ sensitivity are poorly understood. Because the angiotensin II type 1 receptor (AT1R) and the multifunctional transducer protein β-arrestin have been shown to mediate mechanosensitive cellular signaling, we tested the hypothesis that these two proteins are involved in the Frank-Starling mechanism of the heart. Using invasive hemodynamics, we found that mice lacking β-arrestin 1, β-arrestin 2, or AT1R were unable to generate a Frank-Starling force in response to changes in cardiac volume. Although wild-type mice pretreated with the conventional AT1R blocker losartan were unable to enhance cardiac contractility with volume loading, treatment with a β-arrestin-biased AT1R ligand to selectively activate β-arrestin signaling preserved the Frank-Starling relationship. Importantly, in skinned muscle fiber preparations, we found markedly impaired length-dependent myofilament Ca2+ sensitivity in β-arrestin 1, β-arrestin 2, and AT1R knockout mice. Our data reveal β-arrestin 1, β-arrestin 2, and AT1R as key regulatory molecules in the Frank-Starling mechanism, which potentially can be targeted therapeutically with β-arrestin-biased AT1R ligands.
Collapse
|
11
|
Brea MS, Díaz RG, Escudero DS, Caldiz CI, Portiansky EL, Morgan PE, Pérez NG. Epidermal Growth Factor Receptor Silencing Blunts the Slow Force Response to Myocardial Stretch. J Am Heart Assoc 2016; 5:JAHA.116.004017. [PMID: 27744404 PMCID: PMC5121502 DOI: 10.1161/jaha.116.004017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Myocardial stretch increases force biphasically: the Frank‐Starling mechanism followed by the slow force response (SFR). Based on pharmacological strategies, we proposed that epidermal growth factor (EGF) receptor (EGFR or ErbB1) activation is crucial for SFR development. Pharmacological inhibitors could block ErbB4, a member of the ErbB family present in the adult heart. We aimed to specifically test the role of EGFR activation after stretch, with an interference RNA incorporated into a lentiviral vector (small hairpin RNA [shRNA]‐EGFR). Methods and Results Silencing capability of p‐shEGFR was assessed in EGFR‐GFP transiently transfected HEK293T cells. Four weeks after lentivirus injection into the left ventricular wall of Wistar rats, shRNA‐EGFR–injected hearts showed ≈60% reduction of EGFR protein expression compared with shRNA‐SCR–injected hearts. ErbB2 and ErbB4 expression did not change. The SFR to stretch evaluated in isolated papillary muscles was ≈130% of initial rapid phase in the shRNA‐SCR group, while it was blunted in shRNA‐EGFR–expressing muscles. Angiotensin II (Ang II)‐dependent Na+/H+ exchanger 1 activation was indirectly evaluated by intracellular pH measurements in bicarbonate‐free medium, demonstrating an increase in shRNA‐SCR–injected myocardium, an effect not observed in the silenced group. Ang II‐ or EGF‐triggered reactive oxygen species production was significantly reduced in shRNA‐EGFR–injected hearts compared with that in the shRNA‐SCR group. Chronic lentivirus treatment affected neither the myocardial basal redox state (thiobarbituric acid reactive substances) nor NADPH oxidase activity or expression. Finally, Ang II or EGF triggered a redox‐sensitive pathway, leading to p90RSK activation in shRNA‐SCR‐injected myocardium, an effect that was absent in the shRNA‐EGFR group. Conclusions Our results provide evidence that specific EGFR activation after myocardial stretch is a key factor in promoting the redox‐sensitive kinase activation pathway, leading to SFR development.
Collapse
Affiliation(s)
- María S Brea
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Romina G Díaz
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Daiana S Escudero
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia I Caldiz
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Enrique L Portiansky
- Laboratorio de Análisis de Imágenes, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Patricio E Morgan
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Néstor G Pérez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
12
|
Rainer PP, Kass DA. Old dog, new tricks: novel cardiac targets and stress regulation by protein kinase G. Cardiovasc Res 2016; 111:154-62. [PMID: 27297890 DOI: 10.1093/cvr/cvw107] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/18/2016] [Indexed: 12/11/2022] Open
Abstract
The second messenger cyclic guanosine 3'5' monophosphate (cGMP) and its downstream effector protein kinase G (PKG) have been discovered more than 40 years ago. In vessels, PKG1 induces smooth muscle relaxation in response to nitric oxide signalling and thus lowers systemic and pulmonary blood pressure. In platelets, PKG1 stimulation by cGMP inhibits activation and aggregation, and in experimental models of heart failure (HF), PKG1 activation by inhibiting cGMP degradation is protective. The net effect of the above-mentioned signalling is cardiovascular protection. Yet, while modulation of cGMP-PKG has entered clinical practice for treating pulmonary hypertension or erectile dysfunction, translation of promising studies in experimental HF to clinical success has failed thus far. With the advent of new technologies, novel mechanisms of PKG regulation, including mechanosensing, redox regulation, protein quality control, and cGMP degradation, have been discovered. These novel, non-canonical roles of PKG1 may help understand why clinical translation has disappointed thus far. Addressing them appears to be a requisite for future, successful translation of experimental studies to the clinical arena.
Collapse
Affiliation(s)
- Peter P Rainer
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, A-8036 Graz, Austria
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
13
|
Neves JS, Leite-Moreira AM, Neiva-Sousa M, Almeida-Coelho J, Castro-Ferreira R, Leite-Moreira AF. Acute Myocardial Response to Stretch: What We (don't) Know. Front Physiol 2016; 6:408. [PMID: 26779036 PMCID: PMC4700209 DOI: 10.3389/fphys.2015.00408] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/11/2015] [Indexed: 12/24/2022] Open
Abstract
Myocardial stretch, as result of acute hemodynamic overload, is one of the most frequent challenges to the heart and the ability of the heart to intrinsically adapt to it is essential to prevent circulatory congestion. In this review, we highlight the historical background, the currently known mechanisms, as well as the gaps in the understanding of this physiological response. The systolic adaptation to stretch is well-known for over 100 years, being dependent on an immediate increase in contractility—known as the Frank-Starling mechanism—and a further progressive increase—the slow force response. On the other hand, its diastolic counterpart remains largely unstudied. Mechanosensors are structures capable of perceiving mechanical signals and activating pathways that allow their transduction into biochemical responses. Although the connection between these structures and stretch activated pathways remains elusive, we emphasize those most likely responsible for the initiation of the acute response. Calcium-dependent pathways, including angiotensin- and endothelin-related pathways; and cGMP-dependent pathways, comprising the effects of nitric oxide and cardiac natriuretic hormones, embody downstream signaling. The ischemic setting, a paradigmatic situation of acute hemodynamic overload, is also touched upon. Despite the relevant knowledge accumulated, there is much that we still do not know. The quest for further understanding the myocardial response to acute stretch may provide new insights, not only in its physiological importance, but also in the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- João S Neves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - André M Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - Manuel Neiva-Sousa
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - João Almeida-Coelho
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - Ricardo Castro-Ferreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - Adelino F Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| |
Collapse
|
14
|
Mishra S, Bedja D, Amuzie C, Avolio A, Chatterjee S. Prevention of cardiac hypertrophy by the use of a glycosphingolipid synthesis inhibitor in ApoE-/- mice. Biochem Biophys Res Commun 2015; 465:159-64. [PMID: 26253472 DOI: 10.1016/j.bbrc.2015.07.159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 07/31/2015] [Indexed: 01/09/2023]
Abstract
ApoE-/- mice fed a high fat and high cholesterol (HFHC) diet (20% fat and 1.25% cholesterol) from 12 weeks of age to 36 weeks revealed an age-dependent increase in the left ventricular mass (LV mass) and decline in fractional shortening (FS%), which worsened with HFHC diet. These traits are indicative of maladaptive pathological cardiac hypertrophy and dysfunction. This was accompanied by loading of glycosphingolipids and increased gene expression of ANP, BNP in myocardial tissue. Masson's trichrome staining revealed a significant increase in cardiomyocyte size and fibrosis. In contrast, treatment with 5 and 10 μM D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP), an inhibitor of glucosylceramide synthase and lactosylceramide synthase, dose-dependently decreased the load of glycosphingolipids and preserved fractional shortening and maintained left ventricular mass to normal 12-week-old control levels over a 6 month treatment period. Our mechanistic studies showed that D-PDMP inhibited cardiac hypertrophy by inhibiting the phosphorylation of mitogen-activated protein kinase (MAPK). We propose that associating increased glycosphingolipid synthesis with cardiac hypertrophy could serve as a novel approach to prevent this phenotype in experimental animal models of diet -induced atherosclerotic heart disease.
Collapse
Affiliation(s)
- Sumita Mishra
- Johns Hopkins University School of Medicine, Department of Pediatrics and Department of Medicine, Baltimore, USA
| | - Djahida Bedja
- Johns Hopkins University School of Medicine, Department of Cardiology, Baltimore, USA; Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Christine Amuzie
- Johns Hopkins University School of Medicine, Department of Pediatrics and Department of Medicine, Baltimore, USA
| | - Alberto Avolio
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Subroto Chatterjee
- Johns Hopkins University School of Medicine, Department of Pediatrics and Department of Medicine, Baltimore, USA.
| |
Collapse
|
15
|
Ward ML, Shen X, Greenwood DR. Use of liquid chromatography-mass spectrometry (LC-MS) to detect substances of nanomolar concentration in the coronary effluent of isolated perfused hearts. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:270-8. [DOI: 10.1016/j.pbiomolbio.2014.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 01/29/2023]
|
16
|
Ennis IL, Aiello EA, Cingolani HE, Perez NG. The autocrine/paracrine loop after myocardial stretch: mineralocorticoid receptor activation. Curr Cardiol Rev 2014; 9:230-40. [PMID: 23909633 PMCID: PMC3780348 DOI: 10.2174/1573403x113099990034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 12/13/2012] [Indexed: 01/13/2023] Open
Abstract
The stretch of cardiac muscle increases developed force in two phases. The first phase, which occurs rapidly, constitutes the well-known Frank-Starling mechanism and it is generally attributed to enhanced myofilament responsiveness to Ca(2+). The second phase or slow force response (SFR) occurs gradually and is due to an increase in the calcium transient amplitude as a result of a stretch-triggered autocrine/paracrine mechanism. We previously showed that Ca(2+) entry through reverse Na(+)/Ca(2+) exchange underlies the SFR, as the final step of an autocrine/paracrine cascade involving release of angiotensin II/endothelin, and a Na(+)/H(+) exchanger (NHE-1) activation-mediated rise in Na+. In the present review we mainly focus on our three latest contributions to the understanding of this signalling pathway triggered by myocardial stretch: 1) The finding that an increased production of reactive oxygen species (ROS) from mitochondrial origin is critical in the activation of the NHE-1 and therefore in the genesis of the SFR; 2) the demonstration of a key role played by the transactivation of the epidermal growth factor receptor; and 3) the involvement of mineralocorticoid receptors (MR) activation in the stretch-triggered cascade leading to the SFR. Among these novel contributions, the critical role played by the MR is perhaps the most important one. This finding may conceivably provide a mechanistic explanation to the recently discovered strikingly beneficial effects of MR antagonism in humans with cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Irene L Ennis
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Medicas, Universidad Nacional de La Plata, Argentina
| | | | | | | |
Collapse
|
17
|
The signaling pathway for aldosterone-induced mitochondrial production of superoxide anion in the myocardium. J Mol Cell Cardiol 2014; 67:60-8. [DOI: 10.1016/j.yjmcc.2013.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 12/04/2013] [Accepted: 12/08/2013] [Indexed: 01/14/2023]
|
18
|
Correa MV, Nolly MB, Caldiz CI, de Cingolani GEC, Cingolani HE, Ennis IL. Endogenous endothelin 1 mediates angiotensin II-induced hypertrophy in electrically paced cardiac myocytes through EGFR transactivation, reactive oxygen species and NHE-1. Pflugers Arch 2013; 466:1819-30. [PMID: 24327206 DOI: 10.1007/s00424-013-1413-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/26/2013] [Accepted: 11/27/2013] [Indexed: 01/20/2023]
Abstract
Emerging evidence supports a key role for endothelin-1 (ET-1) and the transactivation of the epidermal growth factor receptor (EGFR) in angiotensin II (Ang II) action. We aim to determine the potential role played by endogenous ET-1, EGFR transactivation and redox-dependent sodium hydrogen exchanger-1 (NHE-1) activation in the hypertrophic response to Ang II of cardiac myocytes. Electrically paced adult cat cardiomyocytes were placed in culture and stimulated with 1 nmol l(-1) Ang II or 5 nmol l(-1) ET-1. Ang II increased ~45 % cell surface area (CSA) and ~37 % [(3)H]-phenylalanine incorporation, effects that were blocked not only by losartan (Los) but also by BQ123 (AT1 and ETA receptor antagonists, respectively). Moreover, Ang II significantly increased ET-1 messenger RNA (mRNA) expression. ET-1 similarly increased myocyte CSA and protein synthesis, actions prevented by the reactive oxygen species scavenger MPG or the NHE-1 inhibitor cariporide (carip). ET-1 increased the phosphorylation of the redox-sensitive ERK1/2-p90(RSK) kinases, main activators of the NHE-1. This effect was prevented by MPG and the antagonist of EGFR, AG1478. Ang II, ET-1 and EGF increased myocardial superoxide production (187 ± 9 %, 149 ± 8 % and 163.7 ± 6 % of control, respectively) and AG1478 inhibited these effects. Interestingly, Los inhibited only Ang II whilst BQ123 cancelled both Ang II and ET-1 actions, supporting the sequential and unidirectional activation of AT1, ETA and EGFR. Based on the present evidence, we propose that endogenous ET-1 mediates the hypertrophic response to Ang II by a mechanism that involves EGFR transactivation and redox-dependent activation of the ERK1/2-p90(RSK) and NHE-1 in adult cardiomyocytes.
Collapse
Affiliation(s)
- María V Correa
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120 S/N, La Plata, 1900, Argentina,
| | | | | | | | | | | |
Collapse
|
19
|
Díaz RG, Pérez NG, Morgan PE, Villa-Abrille MC, Caldiz CI, Nolly MB, Portiansky EL, Ennis IL, Cingolani HE. Myocardial mineralocorticoid receptor activation by stretching and its functional consequences. Hypertension 2013; 63:112-8. [PMID: 24126173 DOI: 10.1161/hypertensionaha.113.01726] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Myocardial stretch triggers an angiotensin II-dependent autocrine/paracrine loop of intracellular signals, leading to reactive oxygen species-mediated activation of redox-sensitive kinases. Based on pharmacological strategies, we previously proposed that mineralocorticoid receptor (MR) is necessary for this stretch-triggered mechanism. Now, we aimed to test the role of MR after stretch by using a molecular approach to avoid secondary effects of pharmacological MR blockers. Small hairpin interference RNA capable of specifically knocking down the MR was incorporated into a lentiviral vector (l-shMR) and injected into the left ventricular wall of Wistar rats. The same vector but expressing a nonsilencing sequence (scramble) was used as control. Lentivirus propagation through the left ventricle was evidenced by confocal microscopy. Myocardial MR expression, stretch-triggered activation of redox-sensitive kinases (ERK1/2-p90(RSK)), the consequent Na(+)/H(+) exchanger-mediated changes in pHi (HEPES-buffer), and its mechanical counterpart, the slow force response, were evaluated. Furthermore, reactive oxygen species production in response to a low concentration of angiotensin II (1.0 nmol/L) or an equipotent concentration of epidermal growth factor (0.1 μg/mL) was compared in myocardial tissue slices from both groups. Compared with scramble, animals transduced with l-shMR showed (1) reduced cardiac MR expression, (2) cancellation of angiotensin II-induced reactive oxygen species production but preservation of epidermal growth factor-induced reactive oxygen species production, (3) cancellation of stretch-triggered increase in ERK1/2-p90(RSK) phosphorylation, (4) lack of stretch-induced Na(+)/H(+) exchanger activation, and (5) abolishment of the slow force response. Our results provide strong evidence that MR activation occurs after myocardial stretch and is a key factor to promote redox-sensitive kinase activation and their downstream consequences.
Collapse
Affiliation(s)
- Romina G Díaz
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, UNLP, 60 y 120, 1900 La Plata, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Aiello EA, De Giusti VC. Regulation of the cardiac sodium/bicarbonate cotransporter by angiotensin II: potential Contribution to structural, ionic and electrophysiological myocardial remodelling. Curr Cardiol Rev 2013; 9:24-32. [PMID: 23116057 PMCID: PMC3584305 DOI: 10.2174/157340313805076340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 08/15/2012] [Accepted: 09/13/2012] [Indexed: 12/15/2022] Open
Abstract
The sodium/ bicarbonate cotransporter (NBC) is, with the Na+/H+ exchanger (NHE), an important alkalinizing mechanism that maintains cellular intracellular pH (pHi). In the heart exists at least three isoforms of NBC, one that promotes the co-influx of 1 molecule of Na+ per 1molecule of HCO3-(electroneutral isoform; nNBC) and two others that generates the co-influx of 1 molecule of Na+ per 2 molecules of HCO3- (electrogenic isoforms; eNBC). In addition, the eNBC generates an anionic repolarizing current that modulate the cardiac action potential (CAP), adding to such isoforms the relevance to modulate the electrophysiological function of the heart. Angiotensin II (Ang II) is one of the main hormones that regulate cardiac physiology. The alkalinizing mechanisms (NHE and NBC) are stimulated by Ang II, increasing pHi and intracellular Na+ concentration, which indirectly, due to the stimulation of the Na+/Ca2+ exchanger (NCX) operating in the reverse form, leads to an increase in the intracellular Ca2+ concentration. Interestingly, it has been shown that Ang II exhibits an opposite effect on NBC isoforms: it activates the nNBC and inhibits the eNBC. This inhibition generates a CAP prolongation, which could directly increase the intracellular Ca2+ concentration. The regulation of the intracellular Na+ and Ca2+ concentrations is crucial for the cardiac cellular physiology, but these ions are also involved in the development of cardiac hypertrophy and the damage produced by ischemia-reperfusion, suggesting a potential role of NBC in cardiac diseases.
Collapse
Affiliation(s)
- Ernesto Alejandro Aiello
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, 1900, La Plata, Argentina.
| | | |
Collapse
|
21
|
Shen X, Cannell MB, Ward ML. Effect of SR load and pH regulatory mechanisms on stretch-dependent Ca(2+) entry during the slow force response. J Mol Cell Cardiol 2013; 63:37-46. [PMID: 23880608 DOI: 10.1016/j.yjmcc.2013.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/11/2013] [Accepted: 07/11/2013] [Indexed: 12/22/2022]
Abstract
When cardiac muscle is stretched, there is an initial inotropic response that coincides with the stretch followed by a slower increase in twitch force that develops over several minutes (the "slow force response", or SFR). Unlike the initial response to stretch, the SFR is produced by an increase in Ca(2+) transient amplitude, but the cellular mechanisms that give rise to the increased transients are still debated. We have examined the relationship between the SFR, intracellular [Ca(2+)] and the inotropic state of right ventricular trabeculae from rat hearts at 37°C. The magnitude of the SFR varied with [Ca(2+)]o and stimulation frequency, so that the SFR was greatest for conditions associated with a reduced SR Ca(2+) content. The SFR was not blocked by the AT1 receptor blocker losartan, but was reduced by SN-6, an inhibitor of reverse mode Na(+)/Ca(2+)-exchange (NCX). The Na(+)/H(+)-exchange (NHE) inhibitor HOE642 had no effect in HCO3(-)-buffered solutions, but blocked 50% of the SFR in HCO3(-)-free solution. Inhibition of HCO3(-) transport by DIDS increased the SFR and made it sensitive to HOE642. The addition of cross-bridge cycle inhibitors (20mM BDM or 20μM blebbistatin) to the superfusate reduced the SFR as monitored by changes in Ca(2+). In HCO3(-)-free conditions, the SFR was associated with a slow acidification that was inhibited by BDM, and by stopping electrical stimulation. These results can be explained by stretch increasing metabolic demand and stimulating Na(+) entry via both NHE and the Na(+)/HCO3(-) transporters. This mechanism provides a novel link between inotropic state and stretch, as well as a way for the cell to compensate for increased acid load. The feedback mechanism between force and Ca(2+) transient amplitude that we describe is also limited by the degree of SR Ca(2+) load.
Collapse
Affiliation(s)
- Xin Shen
- Department of Physiology, University of Auckland, Auckland 1023, New Zealand
| | | | | |
Collapse
|
22
|
Alvarez BV, Villa-Abrille MC. Mitochondrial NHE1: a newly identified target to prevent heart disease. Front Physiol 2013; 4:152. [PMID: 23825461 PMCID: PMC3695379 DOI: 10.3389/fphys.2013.00152] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/09/2013] [Indexed: 11/23/2022] Open
Abstract
Mitochondrial damage has been associated with early steps of cardiac dysfunction in heart subjected to ischemic stress, oxidative stress and hypertrophy. A common feature for the mitochondrial deterioration is the loss of the mitochondrial membrane potential (ΔΨ m) with the concomitant irreversible opening of the mitochondrial permeability transition pore (MPTP) which follows the mitochondrial Ca2+ overload, and the subsequent mitochondrial swelling. We have recently characterized the expression of the Na+/H+ exchanger 1 (mNHE1) in mitochondrial membranes. This surprising observation provided a unique target for the prevention of the Ca2+-induced MPTP opening, based on the inhibition of the NHE1 m. In this line, inhibition of NHE1 m activity and/or reduction of NHE1 m expression decreased the Ca2+-induced mitochondrial swelling and the release of reactive oxygen species (ROS) in isolated cardiac mitochondria and preserved the ΔΨ m in isolated cardiomyocytes. Mitochondrial NHE1 thus represents a novel target to prevent cardiac disease, opening new avenues for future research.
Collapse
Affiliation(s)
- Bernardo V Alvarez
- Centro de Investigaciones Cardiovasculares, Consejo Nacional de Investigaciones Científicas y Técnicas Facultad de Ciencias Médicas, Universidad Nacional de La Plata La Plata, Argentina
| | | |
Collapse
|
23
|
De Giusti VC, Caldiz CI, Ennis IL, Pérez NG, Cingolani HE, Aiello EA. Mitochondrial reactive oxygen species (ROS) as signaling molecules of intracellular pathways triggered by the cardiac renin-angiotensin II-aldosterone system (RAAS). Front Physiol 2013; 4:126. [PMID: 23755021 PMCID: PMC3667248 DOI: 10.3389/fphys.2013.00126] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/13/2013] [Indexed: 12/22/2022] Open
Abstract
Mitochondria represent major sources of basal reactive oxygen species (ROS) production of the cardiomyocyte. The role of ROS as signaling molecules that mediate different intracellular pathways has gained increasing interest among physiologists in the last years. In our lab, we have been studying the participation of mitochondrial ROS in the intracellular pathways triggered by the renin-angiotensin II-aldosterone system (RAAS) in the myocardium during the past few years. We have demonstrated that acute activation of cardiac RAAS induces mitochondrial ATP-dependent potassium channel (mitoKATP) opening with the consequent enhanced production of mitochondrial ROS. These oxidant molecules, in turn, activate membrane transporters, as sodium/hydrogen exchanger (NHE-1) and sodium/bicarbonate cotransporter (NBC) via the stimulation of the ROS-sensitive MAPK cascade. The stimulation of such effectors leads to an increase in cardiac contractility. In addition, it is feasible to suggest that a sustained enhanced production of mitochondrial ROS induced by chronic cardiac RAAS, and hence, chronic NHE-1 and NBC stimulation, would also result in the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- V C De Giusti
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, UNLP-CONICET La Plata, Argentina
| | | | | | | | | | | |
Collapse
|
24
|
Vargas LA, Díaz RG, Swenson ER, Pérez NG, Álvarez BV. Inhibition of carbonic anhydrase prevents the Na(+)/H(+) exchanger 1-dependent slow force response to rat myocardial stretch. Am J Physiol Heart Circ Physiol 2013; 305:H228-37. [PMID: 23709596 DOI: 10.1152/ajpheart.00055.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Myocardial stretch is an established signal that leads to hypertrophy. Myocardial stretch induces a first immediate force increase followed by a slow force response (SFR), which is a consequence of an increased Ca(2+) transient that follows the NHE1 Na(+)/H(+) exchanger activation. Carbonic anhydrase II (CAII) binds to the extreme COOH terminus of NHE1 and regulates its transport activity. We aimed to test the role of CAII bound to NHE1 in the SFR. The SFR and changes in intracellular pH (pHi) were evaluated in rat papillary muscle bathed with CO2/HCO3(-) buffer and stretched from 92% to 98% of the muscle maximal force development length for 10 min in the presence of the CA inhibitor 6-ethoxzolamide (ETZ, 100 μM). SFR control was 120 ± 3% (n = 8) of the rapid initial phase and was fully blocked by ETZ (99 ± 4%, n = 6). The SFR corresponded to a maximal increase in pHi of 0.18 ± 0.02 pH units (n = 4), and pHi changes were blocked by ETZ (0.04 ± 0.04, n = 6), as monitored by epifluorescence. NHE1/CAII physical association was examined in the SFR by coimmunoprecipitation, using muscle lysates. CAII immunoprecipitated with an anti-NHE1 antibody and the CAII immunoprecipitated protein levels increased 58 ± 9% (n = 6) upon stretch of muscles, assessed by immunoblots. The p90(RSK) kinase inhibitor SL0101-1 (10 μM) blocked the SFR of heart muscles after stretch 102 ± 2% (n = 4) and reduced the binding of CAII to NHE1, suggesting that the stretch-induced phosphorylation of NHE1 increases its binding to CAII. CAII/NHE1 interaction constitutes a component of the SFR to heart muscle stretch, which potentiates NHE1-mediated H(+) transport in the myocardium.
Collapse
Affiliation(s)
- Lorena A Vargas
- Centro de Investigaciones Cardiovasculares, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | | | | | | | | |
Collapse
|
25
|
Abstract
Myocardial stretch elicits a rapid increase in developed force, which is mainly caused by an increase in myofilament calcium sensitivity (Frank-Starling mechanism). Over the ensuing 10-15 min, a second gradual increase in force takes place. This slow force response to stretch is known to be the result of an increase in the calcium transient amplitude and constitutes the in vitro equivalent of the Anrep effect described 100 years ago in the intact heart. In the present review, we will update and discuss what is known about the Anrep effect as the mechanical counterpart of autocrine/paracrine mechanisms involved in its genesis. The chain of events triggered by myocardial stretch comprises 1) release of angiotensin II, 2) release of endothelin, 3) activation of the mineralocorticoid receptor, 4) transactivation of the epidermal growth factor receptor, 5) increased formation of mitochondria reactive oxygen species, 6) activation of redox-sensitive kinases upstream myocardial Na(+)/H(+) exchanger (NHE1), 7) NHE1 activation, 8) increase in intracellular Na(+) concentration, and 9) increase in Ca(2+) transient amplitude through the Na(+)/Ca(2+) exchanger. We will present the experimental evidence supporting each of the signaling steps leading to the Anrep effect and its blunting by silencing NHE1 expression with a specific small hairpin interference RNA injected into the ventricular wall.
Collapse
Affiliation(s)
- Horacio E Cingolani
- Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, La Plata, Argentina.
| | | | | | | |
Collapse
|
26
|
Caldiz CI, Díaz RG, Nolly MB, Chiappe de Cingolani GE, Ennis IL, Cingolani HE, Pérez NG. Mineralocorticoid receptor activation is crucial in the signalling pathway leading to the Anrep effect. J Physiol 2012; 589:6051-61. [PMID: 22174146 DOI: 10.1113/jphysiol.2011.218750] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The increase in myocardial reactive oxygen species after epidermal growth factor receptor transactivation is a crucial step in the autocrine/paracrine angiotensin II/endothelin receptor activation leading to the slow force response to stretch (SFR). Since experimental evidence suggests a link between angiotensin II or its AT1 receptor and the mineralocorticoid receptor (MR), and MR transactivates the epidermal growth factor receptor, we thought to determine whether MR activation participates in the SFR development in rat myocardium. We show here that MR activation is necessary to promote reactive oxygen species formation by a physiological concentration of angiotensin II (1 nmol l(-1)), since an increase in superoxide anion formation of ~50% of basal was suppressed by blocking MR with spironolactone or eplerenone. This effect was also suppressed by blocking AT1, endothelin (type A) or epidermal growth factor receptors, by inhibiting NADPH oxydase or by targeting mitochondria, and was unaffected by glucocorticoid receptor inhibition. All interventions except AT1 receptor blockade blunted the increase in superoxide anion promoted by an equipotent dose of endothelin-1 (1 nmol l(-1)) confirming that endothelin receptors activation is downstream of AT1. Similarly, an increase in superoxide anion promoted by an equipotent dose of aldosterone (10 nmol l(-1)) was blocked by spironolactone or eplerenone, by preventing epidermal growth factor receptor transactivation, but not by inhibiting glucocorticoid receptors or protein synthesis, suggesting non-genomic MR effects. Combination of aldosterone plus endothelin-1 did not increase superoxide anion formation more than each agonist separately. We found that aldosterone increased phosphorylation of the redox-sensitive kinases ERK1/2-p90RSK and the NHE-1, effects that were eliminated by eplerenone or by preventing epidermal growth factor receptor transactivation. Finally, we provide evidence that the SFR is suppressed by MR blockade, by preventing epidermal growth factor receptor transactivation or by scavenging reactive oxygen species, but it is unaffected by glucocorticoid receptor blockade or protein synthesis inhibition. Our results suggest that MR activation is a necessary step in the stretch-triggered reactive oxygen species-mediated activation of redox-sensitive kinases upstream NHE-1.
Collapse
Affiliation(s)
- Claudia I Caldiz
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | | | | | | | | | | | | |
Collapse
|
27
|
Perjés Á, Kubin A, Kónyi A, Szabados S, Cziráki A, Skoumal R, Ruskoaho H, Szokodi I. Physiological regulation of cardiac contractility by endogenous reactive oxygen species. Acta Physiol (Oxf) 2012; 205:26-40. [PMID: 22463609 DOI: 10.1111/j.1748-1716.2012.02391.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Increased production of reactive oxygen species (ROS) has been linked to the pathogenesis of congestive heart failure. However, emerging evidence suggests the involvement of ROS in the regulation of various physiological cellular processes in the myocardium. In this review, we summarize the latest findings regarding the role of ROS in the acute regulation of cardiac contractility. We discuss ROS-dependent modulation of the inotropic responses to G protein-coupled receptor agonists (e.g. β-adrenergic receptor agonists and endothelin-1), the potential cellular sources of ROS (e.g. NAD(P)H oxidases and mitochondria) and the proposed end-targets and signalling pathways by which ROS affect contractility. Accumulating new data supports the fundamental role of endogenously generated ROS to regulate cardiac function under physiological conditions.
Collapse
Affiliation(s)
| | - A.M. Kubin
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - A. Kónyi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - S. Szabados
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - A. Cziráki
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - R. Skoumal
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - H. Ruskoaho
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - I. Szokodi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| |
Collapse
|
28
|
Perjés Á, Kubin A, Kónyi A, Szabados S, Cziráki A, Skoumal R, Ruskoaho H, Szokodi I. Physiological regulation of cardiac contractility by endogenous reactive oxygen species. Acta Physiol (Oxf) 2012. [DOI: 10.1111/j.1748-1716.2011.02391.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - A.M. Kubin
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - A. Kónyi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - S. Szabados
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - A. Cziráki
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - R. Skoumal
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - H. Ruskoaho
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - I. Szokodi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| |
Collapse
|
29
|
Cingolani OH, Kirk JA, Seo K, Koitabashi N, Lee DI, Ramirez-Correa G, Bedja D, Barth AS, Moens AL, Kass DA. Thrombospondin-4 is required for stretch-mediated contractility augmentation in cardiac muscle. Circ Res 2011; 109:1410-4. [PMID: 22034490 PMCID: PMC3324097 DOI: 10.1161/circresaha.111.256743] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 10/19/2011] [Indexed: 01/26/2023]
Abstract
RATIONALE One of the physiological mechanisms by which the heart adapts to a rise in blood pressure is by augmenting myocyte stretch-mediated intracellular calcium, with a subsequent increase in contractility. This slow force response was first described over a century ago and has long been considered compensatory, but its underlying mechanisms and link to chronic adaptations remain uncertain. Because levels of the matricellular protein thrombospondin-4 (TSP4) rapidly rise in hypertension and are elevated in cardiac stress overload and heart failure, we hypothesized that TSP4 is involved in this adaptive mechanism. OBJECTIVE To determine the mechano-transductive role that TSP4 plays in cardiac regulation to stress. METHODS AND RESULTS In mice lacking TSP4 (Tsp4⁻/⁻), hearts failed to acutely augment contractility or activate stretch-response pathways (ERK1/2 and Akt) on exposure to acute pressure overload. Sustained pressure overload rapidly led to greater chamber dilation, reduced function, and increased heart mass. Unlike controls, Tsp4⁻/⁻ cardiac trabeculae failed to enhance contractility and cellular calcium after a stretch. However, the contractility response was restored in Tsp4⁻/⁻ muscle incubated with recombinant TSP4. Isolated Tsp4⁻/⁻ myocytes responded normally to stretch, identifying a key role of matrix-myocyte interaction for TSP4 contractile modulation. CONCLUSION These results identify TSP4 as myocyte-interstitial mechano-signaling molecule central to adaptive cardiac contractile responses to acute stress, which appears to play a crucial role in the transition to chronic cardiac dilatation and failure.
Collapse
Affiliation(s)
- Oscar H Cingolani
- Division of Cardiology, Department of Medicine, Department of Biomedical Engineering, The Johns Hopkins University Medical Institutions, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
De Giusti VC, Nolly MB, Yeves AM, Caldiz CI, Villa-Abrille MC, Chiappe de Cingolani GE, Ennis IL, Cingolani HE, Aiello EA. Aldosterone Stimulates the Cardiac Na
+
/H
+
Exchanger via Transactivation of the Epidermal Growth Factor Receptor. Hypertension 2011; 58:912-9. [DOI: 10.1161/hypertensionaha.111.176024] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The use of antagonists of the mineralocorticoid receptor in the treatment of myocardial hypertrophy and heart failure has gained increasing importance in the last years. The cardiac Na
+
/H
+
exchanger (NHE-1) upregulation induced by aldosterone could account for the genesis of these pathologies. We tested whether aldosterone-induced NHE-1 stimulation involves the transactivation of the epidermal growth factor receptor (EGFR). Rat ventricular myocytes were used to measure intracellular pH with epifluorescence. Aldosterone enhanced the NHE-1 activity. This effect was canceled by spironolactone or eplerenone (mineralocorticoid receptor antagonists), but not by mifepristone (glucocorticoid receptor antagonist) or cycloheximide (protein synthesis inhibitor), indicating that the mechanism is mediated by the mineralocorticoid receptor triggering nongenomic pathways. Aldosterone-induced NHE-1 stimulation was abolished by the EGFR kinase inhibitor AG1478, suggesting that is mediated by transactivation of EGFR. The increase in the phosphorylation level of the kinase p90
RSK
and NHE-1 serine703 induced by aldosterone was also blocked by AG1478. Exogenous epidermal growth factor mimicked the effects of aldosterone on NHE-1 activity. Epidermal growth factor was also able to increase reactive oxygen species production, and the epidermal growth factor–induced activation of the NHE-1 was abrogated by the reactive oxygen species scavenger
N
-2-mercaptopropionyl glycine, indicating that reactive oxygen species are participating as signaling molecules in this mechanism. Aldosterone enhances the NHE-1 activity via transactivation of the EGFR, formation of reactive oxygen species, and phosphorylation of the exchanger. These results call attention to the consideration of the EGFR as a new potential therapeutic target of the cardiovascular pathologies involving the participation of aldosterone.
Collapse
Affiliation(s)
- Verónica C. De Giusti
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Mariela B. Nolly
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Alejandra M. Yeves
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia I. Caldiz
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María C. Villa-Abrille
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Gladys E. Chiappe de Cingolani
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Irene L. Ennis
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Horacio E. Cingolani
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Ernesto A. Aiello
- From the Centro de Investigaciones Cardiovasculares (CONICET), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
31
|
Pérez NG, Nolly MB, Roldan MC, Villa-Abrille MC, Cingolani E, Portiansky EL, Álvarez BV, Ennis IL, Cingolani HE. Silencing of NHE-1 blunts the slow force response to myocardial stretch. J Appl Physiol (1985) 2011; 111:874-80. [DOI: 10.1152/japplphysiol.01344.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Myocardial stretch induces a biphasic force response: a first abrupt increase followed by a slow force response (SFR), believed to be the in vitro manifestation of the Anrep effect. The SFR is due to an increase in Ca2+ transient of unclear mechanism. We proposed that Na+/H+ exchanger (NHE-1) activation is a key factor in determining the contractile response, but recent reports challenged our findings. We aimed to specifically test the role of the NHE-1 in the SFR. To this purpose small hairpin interference RNA capable of mediating specific NHE-1 knockdown was incorporated into a lentiviral vector (l-shNHE1) and injected into the left ventricular wall of Wistar rats. Injection of a lentiviral vector expressing a nonsilencing sequence (scramble) served as control. Myocardial NHE-1 protein expression and function (the latter evaluated by the recovery of pHi after an acidic load and the SFR) were evaluated. Animals transduced with l-shNHE1 showed reduced NHE-1 expression (45 ± 8% of controls; P < 0.05), and the presence of the lentivirus in the left ventricular myocardium, far from the site of injection, was evidenced by confocal microscopy. These findings correlated with depressed basal pHi recovery after acidosis [maxdpHi/d t 0.055 ± 0.008 (scramble) vs. 0.009 ± 0.004 (l-shNHE1) pH units/min, P < 0.05], leftward shift of the relationship between JH+ (H+ efflux corrected by the intrinsic buffer capacity), and abolishment of SFR (124 ± 2 vs. 101 ± 2% of rapid phase; P < 0.05) despite preserved ERK1/2 phosphorylation [247 ± 12 (stretch) and 263 ± 23 (stretch l-shNHE1) % of control; P < 0.05 vs. nonstretched control], well-known NHE-1 activators. Our results provide strong evidence to propose NHE-1 activation as key factor in determining the SFR to stretch.
Collapse
Affiliation(s)
- Néstor G. Pérez
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Mariela B. Nolly
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Mirian C. Roldan
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María C. Villa-Abrille
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Eugenio Cingolani
- Cedars Sinai Heart Institute, Cedars Sinai Medical Center, Los Angeles, California; and
| | - Enrique L. Portiansky
- Instituto de Patología. Facultad de Cs. Veterinarias, Universidad Nacional de La Plata, La Plata, Argentina
| | - Bernardo V. Álvarez
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Irene L. Ennis
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Horacio E. Cingolani
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
32
|
Cingolani HE, Ennis IL, Aiello EA, Pérez NG. Role of autocrine/paracrine mechanisms in response to myocardial strain. Pflugers Arch 2011; 462:29-38. [DOI: 10.1007/s00424-011-0930-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 01/19/2011] [Accepted: 01/19/2011] [Indexed: 10/18/2022]
|
33
|
Egger M, Domenighetti AA. Adaptive and Maladaptive Remodeling of Cardiomyocyte Excitation-Contraction Coupling by Angiotensin II. Trends Cardiovasc Med 2010; 20:78-85. [DOI: 10.1016/j.tcm.2010.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|