1
|
Ushakov A, Ivanchenko V, Gagarina A. Heart Failure And Type 2 Diabetes Mellitus: Neurohumoral, Histological And Molecular Interconnections. Curr Cardiol Rev 2023; 19:e170622206132. [PMID: 35718961 PMCID: PMC10201898 DOI: 10.2174/1573403x18666220617121144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Heart failure (HF) is a global healthcare burden and a leading cause of morbidity and mortality worldwide. Type 2 diabetes mellitus (T2DM) appears to be one of the major risk factors that significantly worsen HF prognosis and increase the risk of fatal cardiovascular outcomes. Despite a great knowledge of pathophysiological mechanisms involved in HF development and progression, hospitalization rates in patients with HF and concomitant T2DM remain elevated. In this review, we discuss the complex interplay between systemic neurohumoral regulation and local cardiac mechanisms participating in myocardial remodeling and HF development in T2DM with special attention to cardiomyocyte energy metabolism, mitochondrial function and calcium metabolism, cardiomyocyte hypertrophy and death, extracellular matrix remodeling.
Collapse
Affiliation(s)
- A. Ushakov
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - V. Ivanchenko
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| | - A. Gagarina
- Department of Internal Medicine 1, Medical Academy named after S.I. Georgievsky of V.I. Vernadsky Crimean Federal University, Simferopol, Russian Federation
| |
Collapse
|
2
|
Gao H, Yang F, Sattari K, Du X, Fu T, Fu S, Liu X, Lin J, Sun Y, Yao J. Bioinspired two-in-one nanotransistor sensor for the simultaneous measurements of electrical and mechanical cellular responses. SCIENCE ADVANCES 2022; 8:eabn2485. [PMID: 36001656 PMCID: PMC9401615 DOI: 10.1126/sciadv.abn2485] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 07/11/2022] [Indexed: 05/21/2023]
Abstract
The excitation-contraction dynamics in cardiac tissue are the most important physiological parameters for assessing developmental state. We demonstrate integrated nanoelectronic sensors capable of simultaneously probing electrical and mechanical cellular responses. The sensor is configured from a three-dimensional nanotransistor with its conduction channel protruding out of the plane. The structure promotes not only a tight seal with the cell for detecting action potential via field effect but also a close mechanical coupling for detecting cellular force via piezoresistive effect. Arrays of nanotransistors are integrated to realize label-free, submillisecond, and scalable interrogation of correlated cell dynamics, showing advantages in tracking and differentiating cell states in drug studies. The sensor can further decode vector information in cellular motion beyond typical scalar information acquired at the tissue level, hence offering an improved tool for cell mechanics studies. The sensor enables not only improved bioelectronic detections but also reduced invasiveness through the two-in-one converging integration.
Collapse
Affiliation(s)
- Hongyan Gao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Kianoosh Sattari
- Department of Mechanical and Aerospace Engineering, University of Missouri, Columbia, MO 65211, USA
| | - Xian Du
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Tianda Fu
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Shuai Fu
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Xiaomeng Liu
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Jian Lin
- Department of Mechanical and Aerospace Engineering, University of Missouri, Columbia, MO 65211, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Jun Yao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| |
Collapse
|
3
|
Liu Z, Cheng Q, Ma X, Song M. Suppressing Effect of Na +/Ca 2+ Exchanger (NCX) Inhibitors on the Growth of Melanoma Cells. Int J Mol Sci 2022; 23:ijms23020901. [PMID: 35055084 PMCID: PMC8780355 DOI: 10.3390/ijms23020901] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 12/28/2022] Open
Abstract
The role of calcium ion (Ca2+) signaling in tumorigenicity has received increasing attention in melanoma research. Previous Ca2+ signaling studies focused on Ca2+ entry routes, but rarely explored the role of Ca2+ extrusion. Functioning of the Na+/Ca2+ exchanger (NCX) on the plasma membrane is the major way of Ca2+ extrusion, but very few associations between NCX and melanoma have been reported. Here, we explored whether pharmacological modulation of the NCX could suppress melanoma and promise new therapeutic strategies. Methods included cell viability assay, Ca2+ imaging, immunoblotting, and cell death analysis. The NCX inhibitors SN-6 and YM-244769 were used to selectively block reverse operation of the NCX. Bepridil, KB-R7943, and CB-DMB blocked either reverse or forward NCX operation. We found that blocking the reverse NCX with SN-6 or YM-244769 (5–100 μM) did not affect melanoma cells or increase cytosolic Ca2+. Bepridil, KB-R7943, and CB-DMB all significantly suppressed melanoma cells with IC50 values of 3–20 μM. Bepridil and KB-R7943 elevated intracellular Ca2+ level of melanoma. Bepridil-induced melanoma cell death came from cell cycle arrest and enhanced apoptosis, which were all attenuated by the Ca2+ chelator BAPTA-AM. As compared with melanoma, normal melanocytes had lower NCX1 expression and were less sensitive to the cytotoxicity of bepridil. In conclusion, blockade of the forward but not the reverse NCX leads to Ca2+-related cell death in melanoma and the NCX is a potential drug target for cancer therapy.
Collapse
|
4
|
Ottolia M, John S, Hazan A, Goldhaber JI. The Cardiac Na + -Ca 2+ Exchanger: From Structure to Function. Compr Physiol 2021; 12:2681-2717. [PMID: 34964124 DOI: 10.1002/cphy.c200031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Ca2+ homeostasis is essential for cell function and survival. As such, the cytosolic Ca2+ concentration is tightly controlled by a wide number of specialized Ca2+ handling proteins. One among them is the Na+ -Ca2+ exchanger (NCX), a ubiquitous plasma membrane transporter that exploits the electrochemical gradient of Na+ to drive Ca2+ out of the cell, against its concentration gradient. In this critical role, this secondary transporter guides vital physiological processes such as Ca2+ homeostasis, muscle contraction, bone formation, and memory to name a few. Herein, we review the progress made in recent years about the structure of the mammalian NCX and how it relates to function. Particular emphasis will be given to the mammalian cardiac isoform, NCX1.1, due to the extensive studies conducted on this protein. Given the degree of conservation among the eukaryotic exchangers, the information highlighted herein will provide a foundation for our understanding of this transporter family. We will discuss gene structure, alternative splicing, topology, regulatory mechanisms, and NCX's functional role on cardiac physiology. Throughout this article, we will attempt to highlight important milestones in the field and controversial topics where future studies are required. © 2021 American Physiological Society. Compr Physiol 12:1-37, 2021.
Collapse
Affiliation(s)
- Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Scott John
- Department of Medicine (Cardiology), UCLA, Los Angeles, California, USA
| | - Adina Hazan
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
5
|
Korotkov SM, Sobol KV, Shemarova IV, Nesterov VP. Effect of Sodium Ions on Calcium-Loaded Rat Heart Mitochondria and Frog Myocardium. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021060041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
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
|
7
|
Lotteau S, Zhang R, Hazan A, Grabar C, Gonzalez D, Aynaszyan S, Philipson KD, Ottolia M, Goldhaber JI. Acute Genetic Ablation of Cardiac Sodium/Calcium Exchange in Adult Mice: Implications for Cardiomyocyte Calcium Regulation, Cardioprotection, and Arrhythmia. J Am Heart Assoc 2021; 10:e019273. [PMID: 34472363 PMCID: PMC8649274 DOI: 10.1161/jaha.120.019273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Sodium‐calcium (Ca2+) exchanger isoform 1 (NCX1) is the dominant Ca2+ efflux mechanism in cardiomyocytes and is critical to maintaining Ca2+ homeostasis during excitation‐contraction coupling. NCX1 activity has been implicated in the pathogenesis of cardiovascular diseases, but a lack of specific NCX1 blockers complicates experimental interpretation. Our aim was to develop a tamoxifen‐inducible NCX1 knockout (KO) mouse to investigate compensatory adaptations of acute ablation of NCX1 on excitation‐contraction coupling and intracellular Ca2+ regulation, and to examine whether acute KO of NCX1 confers resistance to triggered arrhythmia and ischemia/reperfusion injury. Methods and Results We used the α‐myosin heavy chain promoter (Myh6)‐MerCreMer promoter to create a tamoxifen‐inducible cardiac‐specific NCX1 KO mouse. Within 1 week of tamoxifen injection, NCX1 protein expression and current were dramatically reduced. Diastolic Ca2+ increased despite adaptive reductions in Ca2+ current and action potential duration and compensatory increases in excitation‐contraction coupling gain, sarcoplasmic reticulum Ca2+ ATPase 2 and plasma membrane Ca2+ ATPase. As these adaptations progressed over 4 weeks, diastolic Ca2+ normalized and SR Ca2+ load increased. Left ventricular function remained normal, but mild fibrosis and hypertrophy developed. Transcriptomics revealed modification of cardiovascular‐related gene networks including cell growth and fibrosis. NCX1 KO reduced spontaneous action potentials triggered by delayed afterdepolarizations and reduced scar size in response to ischemia/reperfusion. Conclusions Tamoxifen‐inducible NCX1 KO mice adapt to acute genetic ablation of NCX1 by reducing Ca2+ influx, increasing alternative Ca2+ efflux pathways, and increasing excitation‐contraction coupling gain to maintain contractility at the cost of mild Ca2+‐activated hypertrophy and fibrosis and decreased survival. Nevertheless, KO myocytes are protected against spontaneous action potentials and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Sabine Lotteau
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Rui Zhang
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Adina Hazan
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Christina Grabar
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Devina Gonzalez
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | | | - Kenneth D Philipson
- Department of Physiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Michela Ottolia
- Division of Molecular Medicine Department of Anesthesiology and Perioperative Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| | | |
Collapse
|
8
|
Cardiovascular toxicity of PI3Kα inhibitors. Clin Sci (Lond) 2021; 134:2595-2622. [PMID: 33063821 DOI: 10.1042/cs20200302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
The phosphoinositide 3-kinases (PI3Ks) are a family of intracellular lipid kinases that phosphorylate the 3'-hydroxyl group of inositol membrane lipids, resulting in the production of phosphatidylinositol 3,4,5-trisphosphate from phosphatidylinositol 4,5-bisphosphate. This results in downstream effects, including cell growth, proliferation, and migration. The heart expresses three PI3K class I enzyme isoforms (α, β, and γ), and these enzymes play a role in cardiac cellular survival, myocardial hypertrophy, myocardial contractility, excitation, and mechanotransduction. The PI3K pathway is associated with various disease processes but is particularly important to human cancers since many gain-of-function mutations in this pathway occur in various cancers. Despite the development, testing, and regulatory approval of PI3K inhibitors in recent years, there are still significant challenges when creating and utilizing these drugs, including concerns of adverse effects on the heart. There is a growing body of evidence from preclinical studies revealing that PI3Ks play a crucial cardioprotective role, and thus inhibition of this pathway could lead to cardiac dysfunction, electrical remodeling, vascular damage, and ultimately, cardiovascular disease. This review will focus on PI3Kα, including the mechanisms underlying the adverse cardiovascular effects resulting from PI3Kα inhibition and the potential clinical implications of treating patients with these drugs, such as increased arrhythmia burden, biventricular cardiac dysfunction, and impaired recovery from cardiotoxicity. Recommendations for future directions for preclinical and clinical work are made, highlighting the possible role of PI3Kα inhibition in the progression of cancer-related cachexia and female sex and pre-existing comorbidities as independent risk factors for cardiac abnormalities after cancer treatment.
Collapse
|
9
|
Pelat M, Barbe F, Daveu C, Ly-Nguyen L, Lartigue T, Marque S, Tavares G, Ballet V, Guillon JM, Steinmeyer K, Wirth K, Gögelein H, Arndt P, Rackelmann N, Weston J, Bellevergue P, McCort G, Trellu M, Lucats L, Beauverger P, Pruniaux-Harnist MP, Janiak P, Chézalviel-Guilbert F. SAR340835, a Novel Selective Na +/Ca 2+ Exchanger Inhibitor, Improves Cardiac Function and Restores Sympathovagal Balance in Heart Failure. J Pharmacol Exp Ther 2021; 377:293-304. [PMID: 33602875 DOI: 10.1124/jpet.120.000238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/08/2021] [Indexed: 11/22/2022] Open
Abstract
In failing hearts, Na+/Ca2+ exchanger (NCX) overactivity contributes to Ca2+ depletion, leading to contractile dysfunction. Inhibition of NCX is expected to normalize Ca2+ mishandling, to limit afterdepolarization-related arrhythmias, and to improve cardiac function in heart failure (HF). SAR340835/SAR296968 is a selective NCX inhibitor for all NCX isoforms across species, including human, with no effect on the native voltage-dependent calcium and sodium currents in vitro. Additionally, it showed in vitro and in vivo antiarrhythmic properties in several models of early and delayed afterdepolarization-related arrhythmias. Its effect on cardiac function was studied under intravenous infusion at 250,750 or 1500 µg/kg per hour in dogs, which were either normal or submitted to chronic ventricular pacing at 240 bpm (HF dogs). HF dogs were infused with the reference inotrope dobutamine (10 µg/kg per minute, i.v.). In normal dogs, NCX inhibitor increased cardiac contractility (dP/dtmax) and stroke volume (SV) and tended to reduce heart rate (HR). In HF dogs, NCX inhibitor significantly and dose-dependently increased SV from the first dose (+28.5%, +48.8%, and +62% at 250, 750, and 1500 µg/kg per hour, respectively) while significantly increasing dP/dtmax only at 1500 (+33%). Furthermore, NCX inhibitor significantly restored sympathovagal balance and spontaneous baroreflex sensitivity (BRS) from the first dose and reduced HR at the highest dose. In HF dogs, dobutamine significantly increased dP/dtmax and SV (+68.8%) but did not change HR, sympathovagal balance, or BRS. Overall, SAR340835, a selective potent NCX inhibitor, displayed a unique therapeutic profile, combining antiarrhythmic properties, capacity to restore systolic function, sympathovagal balance, and BRS in HF dogs. NCX inhibitors may offer new therapeutic options for acute HF treatment. SIGNIFICANCE STATEMENT: HF is facing growing health and economic burden. Moreover, patients hospitalized for acute heart failure are at high risk of decompensation recurrence, and no current acute decompensated HF therapy definitively improved outcomes. A new potent, Na+/Ca2+ exchanger inhibitor SAR340835 with antiarrhythmic properties improved systolic function of failing hearts without creating hypotension, while reducing heart rate and restoring sympathovagal balance. SAR340835 may offer a unique and attractive pharmacological profile for patients with acute heart failure as compared with current inotrope, such as dobutamine.
Collapse
Affiliation(s)
- Michel Pelat
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Fabrice Barbe
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Cyril Daveu
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Laetitia Ly-Nguyen
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Thomas Lartigue
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Suzanne Marque
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Georges Tavares
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Véronique Ballet
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Jean-Michel Guillon
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Klaus Steinmeyer
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Klaus Wirth
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Heinz Gögelein
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Petra Arndt
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Nils Rackelmann
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - John Weston
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Patrice Bellevergue
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Gary McCort
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Marc Trellu
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Laurence Lucats
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Philippe Beauverger
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Marie-Pierre Pruniaux-Harnist
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Philip Janiak
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| | - Frédérique Chézalviel-Guilbert
- Cardiovascular and Metabolism TSU (M.P., F.B., C.D., T.L., S.M., G.T., L.L., Ph.B., M.-P.P.-H., P.J., F.C.-G.) and Integrated Drug Discovery (Pa.B.), Sanofi R&D, Chilly Mazarin, France; Preclinical Safety, Sanofi R&D, Alfortville, France (L.L.-N., V.B., J.-M.G., M.T.); Sanofi R&D, Industriepark Höchst, Frankfurt, Germany (K.S., K.W., H.G., P.A., N.R., J.W.); and Integrated Drug Discovery, Sanofi R&D, Vitry sur Seine, France (G.M.)
| |
Collapse
|
10
|
Izumi Y, Mennerick SJ, Doherty JJ, Zorumski CF. Oxysterols Modulate the Acute Effects of Ethanol on Hippocampal N-Methyl-d-Aspartate Receptors, Long-Term Potentiation, and Learning. J Pharmacol Exp Ther 2021; 377:181-188. [PMID: 33441369 PMCID: PMC8051516 DOI: 10.1124/jpet.120.000376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/08/2021] [Indexed: 12/29/2022] Open
Abstract
Ethanol is a noncompetitive inhibitor of N-methyl-d-aspartate receptors (NMDARs) and acutely disrupts hippocampal synaptic plasticity and learning. In the present study, we examined the effects of oxysterol positive allosteric modulators (PAMs) of NMDARs on ethanol-mediated inhibition of NMDARs, block of long-term potentiation (LTP) and long-term depression (LTD) in rat hippocampal slices, and defects in one-trial learning in vivo. We found that 24S-hydroxycholesterol and a synthetic oxysterol analog, SGE-301, overcame effects of ethanol on NMDAR-mediated synaptic responses in the CA1 region but did not alter acute effects of ethanol on LTD; the synthetic oxysterol, however, overcame acute inhibition of LTP. In addition, both oxysterols overcame persistent effects of ethanol on LTP in vitro, and the synthetic analog reversed defects in one-trial inhibitory avoidance learning in vivo. These results indicate that effects of ethanol on both LTP and LTD arise by complex mechanisms beyond NMDAR antagonism and that oxysterol NMDAR PAMS may represent a novel approach for preventing and reversing acute ethanol-mediated changes in cognition.
Collapse
Affiliation(s)
- Yukitoshi Izumi
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| | - Steven J Mennerick
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| | - James J Doherty
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| | - Charles F Zorumski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri (Y.I., S.J.M., C.F.Z.); and Sage Therapeutics, Cambridge, Massachusetts (J.J.D.)
| |
Collapse
|
11
|
King DR, Padget RL, Perry J, Hoeker G, Smyth JW, Brown DA, Poelzing S. Elevated perfusate [Na +] increases contractile dysfunction during ischemia and reperfusion. Sci Rep 2020; 10:17289. [PMID: 33057157 PMCID: PMC7560862 DOI: 10.1038/s41598-020-74069-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Recent studies revealed that relatively small changes in perfusate sodium ([Na+]o) composition significantly affect cardiac electrical conduction and stability in contraction arrested ex vivo Langendorff heart preparations before and during simulated ischemia. Additionally, [Na+]o modulates cardiomyocyte contractility via a sodium-calcium exchanger (NCX) mediated pathway. It remains unknown, however, whether modest changes to [Na+]o that promote electrophysiologic stability similarly improve mechanical function during baseline and ischemia-reperfusion conditions. The purpose of this study was to quantify cardiac mechanical function during ischemia-reperfusion with perfusates containing 145 or 155 mM Na+ in Langendorff perfused isolated rat heart preparations. Relative to 145 mM Na+, perfusion with 155 mM [Na+]o decreased the amplitude of left-ventricular developed pressure (LVDP) at baseline and accelerated the onset of ischemic contracture. Inhibiting NCX with SEA0400 abolished LVDP depression caused by increasing [Na+]o at baseline and reduced the time to peak ischemic contracture. Ischemia-reperfusion decreased LVDP in all hearts with return of intrinsic activity, and reperfusion with 155 mM [Na+]o further depressed mechanical function. In summary, elevating [Na+]o by as little as 10 mM can significantly modulate mechanical function under baseline conditions, as well as during ischemia and reperfusion. Importantly, clinical use of Normal Saline, which contains 155 mM [Na+]o, with cardiac ischemia may require further investigation.
Collapse
Affiliation(s)
- D Ryan King
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Rachel L Padget
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Justin Perry
- Department of Human Nutrition, Virginia Polytechnic Institute and State University, Foods, and Exercise, Blacksburg, VA, USA
| | - Gregory Hoeker
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - James W Smyth
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - David A Brown
- Department of Human Nutrition, Virginia Polytechnic Institute and State University, Foods, and Exercise, Blacksburg, VA, USA
| | - Steven Poelzing
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA. .,Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA. .,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA. .,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
12
|
Scranton K, John S, Escobar A, Goldhaber JI, Ottolia M. Modulation of the cardiac Na +-Ca 2+ exchanger by cytoplasmic protons: Molecular mechanisms and physiological implications. Cell Calcium 2019; 87:102140. [PMID: 32070924 DOI: 10.1016/j.ceca.2019.102140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/06/2019] [Accepted: 12/07/2019] [Indexed: 01/31/2023]
Abstract
A precise temporal and spatial control of intracellular Ca2+ concentration is essential for a coordinated contraction of the heart. Following contraction, cardiac cells need to rapidly remove intracellular Ca2+ to allow for relaxation. This task is performed by two transporters: the plasma membrane Na+-Ca2+ exchanger (NCX) and the sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA). NCX extrudes Ca2+ from the cell, balancing the Ca2+entering the cytoplasm during systole through L-type Ca2+ channels. In parallel, following SR Ca2+ release, SERCA activity replenishes the SR, reuptaking Ca2+ from the cytoplasm. The activity of the mammalian exchanger is fine-tuned by numerous ionic allosteric regulatory mechanisms. Micromolar concentrations of cytoplasmic Ca2+ potentiate NCX activity, while an increase in intracellular Na+ levels inhibits NCX via a mechanism known as Na+-dependent inactivation. Protons are also powerful inhibitors of NCX activity. By regulating NCX activity, Ca2+, Na+ and H+ couple cell metabolism to Ca2+ homeostasis and therefore cardiac contractility. This review summarizes the recent progress towards the understanding of the molecular mechanisms underlying the ionic regulation of the cardiac NCX with special emphasis on pH modulation and its physiological impact on the heart.
Collapse
Affiliation(s)
- Kyle Scranton
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Scott John
- Department of Medicine (Cardiology), UCLA, Los Angeles, CA 90095, USA; Cardiovascular Research Laboratory, UCLA, Los Angeles, CA 90095, USA
| | - Ariel Escobar
- Department of Bioengineering, School of Engineering, UC Merced, Merced, CA 95343, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, UCLA, Los Angeles, CA 90095, USA; Cardiovascular Research Laboratory, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
13
|
Wang Z, Ye D, Ye J, Wang M, Liu J, Jiang H, Xu Y, Zhang J, Chen J, Wan J. The TRPA1 Channel in the Cardiovascular System: Promising Features and Challenges. Front Pharmacol 2019; 10:1253. [PMID: 31680989 PMCID: PMC6813932 DOI: 10.3389/fphar.2019.01253] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
The transient receptor potential ankyrin 1 (TRPA1) channel is a calcium-permeable nonselective cation channel in the plasma membrane that belongs to the transient receptor potential (TRP) channel superfamily. Recent studies have suggested that the TRPA1 channel plays an essential role in the development and progression of several cardiovascular conditions, such as atherosclerosis, heart failure, myocardial ischemia-reperfusion injury, myocardial fibrosis, arrhythmia, vasodilation, and hypertension. Activation of the TRPA1 channel has a protective effect against the development of atherosclerosis. Furthermore, TRPA1 channel activation elicits peripheral vasodilation and induces a biphasic blood pressure response. However, loss of channel expression or blockade of its activation suppressed heart failure, myocardial ischemia-reperfusion injury, myocardial fibrosis, and arrhythmia. In this paper, we review recent research progress on the TRPA1 channel and discuss its potential role in the cardiovascular system.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huimin Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jiangbin Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
14
|
Liskova V, Hudecova S, Lencesova L, Iuliano F, Sirova M, Ondrias K, Pastorekova S, Krizanova O. Type 1 Sodium Calcium Exchanger Forms a Complex with Carbonic Anhydrase IX and Via Reverse Mode Activity Contributes to pH Control in Hypoxic Tumors. Cancers (Basel) 2019; 11:cancers11081139. [PMID: 31395807 PMCID: PMC6721473 DOI: 10.3390/cancers11081139] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/02/2019] [Accepted: 08/07/2019] [Indexed: 12/30/2022] Open
Abstract
Hypoxia and acidosis are among the key microenvironmental factors that contribute to cancer progression. We have explored a possibility that the type 1Na+/Ca2+ exchanger (NCX1) is involved in pH control in hypoxic tumors. We focused on changes in intracellular pH, co-localization of NCX1, carbonic anhydrase IX (CA IX), and sodium proton exchanger type 1 (NHE1) by proximity ligation assay, immunoprecipitation, spheroid formation assay and migration of cells due to treatment with KB-R7943, a selective inhibitor of the reverse-mode NCX1. In cancer cells exposed to hypoxia, reverse-mode NCX1 forms a membrane complex primarily with CA IX and also with NHE1. NCX1/CA IX/NHE1 assembly operates as a metabolon with a potent ability to extrude protons to the extracellular space and thereby facilitate acidosis. KB-R7943 prevents formation of this metabolon and reduces cell migration. Thus, we have shown that in hypoxic cancer cells, NCX1 operates in a reverse mode and participates in pH regulation in hypoxic tumors via cooperation with CAIX and NHE1.
Collapse
Affiliation(s)
- Veronika Liskova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Sona Hudecova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Lubomira Lencesova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Filippo Iuliano
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Marta Sirova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Karol Ondrias
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Silvia Pastorekova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia
| | - Olga Krizanova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta, 84505 Bratislava, Slovakia.
- Department of Chemistry, Faculty of Natural Sciences, University of Ss. Cyril and Methodius, Námestie J. Herdu 2, Trnava, 91701 Slovakia.
| |
Collapse
|
15
|
Szadvari I, Hudecova S, Chovancova B, Matuskova M, Cholujova D, Lencesova L, Valerian D, Ondrias K, Babula P, Krizanova O. Sodium/calcium exchanger is involved in apoptosis induced by H 2S in tumor cells through decreased levels of intracellular pH. Nitric Oxide 2019; 87:1-9. [PMID: 30849492 DOI: 10.1016/j.niox.2019.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/31/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023]
Abstract
We explored possibility that sodium/calcium exchanger 1 (NCX1) is involved in pH modulation and apoptosis induction in GYY4137 treated cells. We have shown that although 10 days treatment with GYY4137 did not significantly decreased volume of tumors induced by colorectal cancer DLD1 cells in nude mice, it already induced apoptosis in these tumors. Treatment of DLD1 and ovarian cancer A2780 cells with GYY4137 resulted in intracellular acidification in a concentration-dependent manner. We observed increased mRNA and protein expression of both, NCX1 and sodium/hydrogen exchanger 1 (NHE1) in DLD1-induced tumors from GYY4137-treated mice. NCX1 was coupled with NHE1 in A2780 and DLD1 cells and this complex partially disintegrated after GYY4137 treatment. We proposed that intracellular acidification is due to uncoupling of NCX1/NHE1 complex rather than blocking of the reverse mode of NCX1, probably due to internalization of NHE1. Results might contribute to understanding molecular mechanism of H2S-induced apoptosis in tumor cells.
Collapse
Affiliation(s)
- Ivan Szadvari
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sona Hudecova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbora Chovancova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Miroslava Matuskova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Dana Cholujova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubomira Lencesova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - David Valerian
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Karol Ondrias
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Olga Krizanova
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
16
|
Chu L, Greenstein JL, Winslow RL. Na + microdomains and sparks: Role in cardiac excitation-contraction coupling and arrhythmias in ankyrin-B deficiency. J Mol Cell Cardiol 2019; 128:145-157. [PMID: 30731085 DOI: 10.1016/j.yjmcc.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 02/01/2019] [Accepted: 02/02/2019] [Indexed: 01/25/2023]
Abstract
Cardiac sodium (Na+) potassium ATPase (NaK) pumps, neuronal sodium channels (INa), and sodium calcium (Ca2+) exchangers (NCX1) may co-localize to form a Na+ microdomain. It remains controversial as to whether neuronal INa contributes to local Na+ accumulation, resulting in reversal of nearby NCX1 and influx of Ca2+ into the cell. Therefore, there has been great interest in the possible roles of a Na+ microdomain in cardiac Ca2+-induced Ca2+ release (CICR). In addition, the important role of co-localization of NaK and NCX1 in regulating localized Na+ and Ca2+ levels and CICR in ankyrin-B deficient (ankyrin-B+/-) cardiomyocytes has been examined in many recent studies. Altered Na+ dynamics may contribute to the appearance of arrhythmias, but the mechanisms underlying this relationship remain unclear. In order to investigate this, we present a mechanistic canine cardiomyocyte model which reproduces independent local dyadic junctional SR (JSR) Ca2+ release events underlying cell-wide excitation-contraction coupling, as well as a three-dimensional super-resolution model of the Ca2+ spark that describes local Na+ dynamics as governed by NaK pumps, neuronal INa, and NCX1. The model predicts the existence of Na+ sparks, which are generated by NCX1 and exhibit significantly slower dynamics as compared to Ca2+ sparks. Moreover, whole-cell simulations indicate that neuronal INa in the cardiac dyad plays a key role during the systolic phase. Rapid inward neuronal INa can elevate dyadic [Na+] to 35-40 mM, which drives reverse-mode NCX1 transport, and therefore promotes Ca2+ entry into the dyad, enhancing the trigger for JSR Ca2+ release. The specific role of decreased co-localization of NaK and NCX1 in ankyrin-B+/- cardiomyocytes was examined. Model results demonstrate that a reduction in the local NCX1- and NaK-mediated regulation of dyadic [Ca2+] and [Na+] results in an increase in Ca2+ spark activity during isoproterenol stimulation, which in turn stochastically activates NCX1 in the dyad. This alteration in NCX1/NaK co-localization interrupts the balance between NCX1 and NaK currents in a way that leads to enhanced depolarizing inward current during the action potential plateau, which ultimately leads to a higher probability of L-type Ca2+ channel reopening and arrhythmogenic early-afterdepolarizations.
Collapse
Affiliation(s)
- Lulu Chu
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD 21218, USA.
| | - Joseph L Greenstein
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD 21218, USA.
| | - Raimond L Winslow
- Department of Biomedical Engineering and the Institute for Computational Medicine, The Johns Hopkins University School of Medicine and Whiting School of Engineering, 3400 N Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
17
|
Role of the TRPM4 Channel in Cardiovascular Physiology and Pathophysiology. Cells 2018; 7:cells7060062. [PMID: 29914130 PMCID: PMC6025450 DOI: 10.3390/cells7060062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 12/31/2022] Open
Abstract
The transient receptor potential cation channel subfamily M member 4 (TRPM4) channel influences calcium homeostasis during many physiological activities such as insulin secretion, immune response, respiratory reaction, and cerebral vasoconstriction. This calcium-activated, monovalent, selective cation channel also plays a key role in cardiovascular pathophysiology; for example, a mutation in the TRPM4 channel leads to cardiac conduction disease. Recently, it has been suggested that the TRPM4 channel is also involved in the development of cardiac ischemia-reperfusion injury, which causes myocardial infarction. In the present review, we discuss the physiological function of the TRPM4 channel, and assess its role in cardiovascular pathophysiology.
Collapse
|
18
|
Bai XJ, Hao JT, Wang J, Zhang WF, Yan CP, Zhao JH, Zhao ZQ. Curcumin inhibits cardiac hypertrophy and improves cardiovascular function via enhanced Na +/Ca 2+ exchanger expression after transverse abdominal aortic constriction in rats. Pharmacol Rep 2017; 70:60-68. [PMID: 29331788 DOI: 10.1016/j.pharep.2017.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 07/08/2017] [Accepted: 07/13/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND This study tested the hypothesis that inhibition of cardiac hypertrophy and preservation of cardiac/endothelial function by the natural yellow pigment curcumin are associated with upregulated expression of Na+/Ca2+ exchanger (NCX) after transverse aortic constriction (TAC). METHODS Male Wistar rats were subjected to TAC for 10 weeks and curcumin (50 mg/kg/day) was fed by gastric gavage during TAC. Expression of NCX and endothelial nitric oxide synthase (eNOS) was analyzed by Western blot and immunohistochemistry. RESULTS Compared with the animals in the TAC group, curcumin significantly increased the survival rate and reduced the ratio of heart or left ventricle (LV) to body weight and the cross sectional area of cardiomyocytes. In coincidence with improved LV systolic pressure and reduced LV end-diastolic pressure, curcumin significantly reduced LV end-systolic and diastolic diameter/dimension, and enhanced LV ejection fraction and LV fractional shortening as measured by echocardiography. Furthermore, endothelium-dependent relaxation of aortic rings in response to acetylcholine was significantly improved by curcumin. Along with these modifications, the expression and localization of NCX and eNOS in the myocardium and vascular endothelium were significantly upregulated by curcumin. The protective effect of curcumin on endothelium-dependent relaxation was partly blocked by pretreatment with the NCX inhibitor, KB-R7943. CONCLUSIONS These results demonstrate that inhibition of cardiac hypertrophy, improvement of cardiac systolic/diastolic function and preservation of vascular endothelium by curcumin might be associated with upregulated NCX expression level in response to increased afterload.
Collapse
Affiliation(s)
- Xiao-Jie Bai
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
| | - Jun-Tao Hao
- Department of Thoracic Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, People's Republic of China
| | - Jin Wang
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Wei-Fang Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Cai-Ping Yan
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Jia-Hui Zhao
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Zhi-Qing Zhao
- Cardiovascular Research Laboratory, Mercer University School of Medicine, Savannah, GA, USA
| |
Collapse
|
19
|
Different Densities of Na-Ca Exchange Current in T-Tubular and Surface Membranes and Their Impact on Cellular Activity in a Model of Rat Ventricular Cardiomyocyte. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6343821. [PMID: 28321411 PMCID: PMC5340987 DOI: 10.1155/2017/6343821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/18/2016] [Accepted: 01/04/2017] [Indexed: 01/13/2023]
Abstract
The ratio of densities of Na-Ca exchanger current (INaCa) in the t-tubular and surface membranes (INaCa-ratio) computed from the values of INaCa and membrane capacitances (Cm) measured in adult rat ventricular cardiomyocytes before and after detubulation ranges between 1.7 and 25 (potentially even 40). Variations of action potential waveform and of calcium turnover within this span of the INaCa-ratio were simulated employing previously developed model of rat ventricular cell incorporating separate description of ion transport systems in the t-tubular and surface membranes. The increase of INaCa-ratio from 1.7 to 25 caused a prolongation of APD (duration of action potential at 90% repolarisation) by 12, 9, and 6% and an increase of peak intracellular Ca2+ transient by 45, 19, and 6% at 0.1, 1, and 5 Hz, respectively. The prolonged APD resulted from the increase of INaCa due to the exposure of a larger fraction of Na-Ca exchangers to higher Ca2+ transients under the t-tubular membrane. The accompanying rise of Ca2+ transient was a consequence of a higher Ca2+ load in sarcoplasmic reticulum induced by the increased Ca2+ cycling between the surface and t-tubular membranes. However, the reason for large differences in the INaCa-ratio assessed from measurements in adult rat cardiomyocytes remains to be explained.
Collapse
|
20
|
Zhang YH. Neuronal nitric oxide synthase in hypertension - an update. Clin Hypertens 2016; 22:20. [PMID: 27822383 PMCID: PMC5093926 DOI: 10.1186/s40885-016-0055-8] [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: 07/15/2016] [Accepted: 10/19/2016] [Indexed: 02/07/2023] Open
Abstract
Hypertension is a prevalent condition worldwide and is the key risk factor for fatal cardiovascular complications, such as stroke, sudden cardiac death and heart failure. Reduced bioavailability of nitric oxide (NO) in the endothelium is an important precursor for impaired vasodilation and hypertension. In the heart, NO deficiency deteriorates the adverse consequences of pressure-overload and causes cardiac hypertrophy, fibrosis and myocardial infarction which lead to fatal heart failure and sudden cardiac death. Recent consensus is that both endothelial and neuronal nitric oxide synthases (eNOS or NOS3 and nNOS or NOS1) are the constitutive sources of NO in the myocardium. Between the two, nNOS is the predominant isoform of NOS that controls intracellular Ca2+ homeostasis, myocyte contraction, relaxation and signaling pathways including nitroso-redox balance. Notably, our recent research indicates that cardiac eNOS protein is reduced but nNOS protein expression and activity are increased in hypertension. Furthermore, nNOS is induced by the interplay between angiotensin II (Ang II) type 1 receptor (AT1R) and Ang II type 2 receptor (AT2R), mediated by NADPH oxidase and reactive oxygen species (ROS)-dependent eNOS activity in cardiac myocytes. nNOS, in turn, protects the heart from pathogenesis via positive lusitropy in hypertension. Soluble guanylate cyclase (sGC)-cGMP/PKG-dependent phosphorylation of myofilament proteins are novel targets of nNOS in hypertensive myocardium. In this short review, we will endeavor to overview new findings of the up-stream and downstream regulation of cardiac nNOS in hypertension, shed light on the underlying mechanisms which may be of therapeutic value in hypertensive cardiomyopathy.
Collapse
Affiliation(s)
- Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, 103 Dae Hak Ro, Chong No Gu, 110-799 Seoul Korea ; Yanbian University Hospital, Yanji, Jilin Province 133000 China ; Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Grover AK. Sodium-Calcium Exchanger in Pig Coronary Artery. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:145-170. [PMID: 28212796 DOI: 10.1016/bs.apha.2016.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review focuses on the sodium-calcium exchangers (NCX) in the left anterior descending coronary artery smooth muscle. Bathing tissues in Na+-substituted solutions caused them to contract. In cultured smooth muscle cells, it increased the cytosolic Ca2+ concentration and extracellular entry of 45Ca2+. All three activities were attributed to NCX since they were inhibited by NCX inhibitors. The tissues also expressed the sarco/endoplasmic reticulum (SER) Ca2+ pump SERCA2b whose activity was much greater than that of NCX. Inhibiting SERCA2b with thapsigargin decreased the NCX-mediated 45Ca2+ accumulation by the cells. The decrease was not observed in cells loaded with the Ca2+-chelator BAPTA. The results are consistent with a limited diffusional space model with a proximity between NCX and SERCA2b. NCX molecules appear to be colocalized with the subsarcolemmal SERCA2b based on studies on membrane flotation experiments and microscopic fluorescence imaging of antibody-labeled cells. Thapsigargin inhibition of SERCA2b moved NCX even closer to SER. This provides a model for the NCX-mediated Ca2+ refilling of SER in the arterial smooth muscle. The model for the NCX-mediated refilling of the depleted SER proposed for smooth muscle did not apply to endothelium in which NCX levels were greater and SERCA levels were lower than in smooth muscle. The effect of thapsigargin on the NCX-mediated Ca2+ accumulation which was observed in smooth muscle was absent in the endothelium. We propose that the coupling between NCX and smooth muscle may be tissue dependent.
Collapse
Affiliation(s)
- A K Grover
- McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
22
|
Smith SA, Hughes LD, Kline CF, Kempton AN, Dorn LE, Curran J, Makara M, Webb TR, Wright P, Voigt N, Binkley PF, Janssen PML, Kilic A, Carnes CA, Dobrev D, Rasband MN, Hund TJ, Mohler PJ. Dysfunction of the β2-spectrin-based pathway in human heart failure. Am J Physiol Heart Circ Physiol 2016; 310:H1583-91. [PMID: 27106045 DOI: 10.1152/ajpheart.00875.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/11/2016] [Indexed: 11/22/2022]
Abstract
β2-Spectrin is critical for integrating membrane and cytoskeletal domains in excitable and nonexcitable cells. The role of β2-spectrin for vertebrate function is illustrated by dysfunction of β2-spectrin-based pathways in disease. Recently, defects in β2-spectrin association with protein partner ankyrin-B were identified in congenital forms of human arrhythmia. However, the role of β2-spectrin in common forms of acquired heart failure and arrhythmia is unknown. We report that β2-spectrin protein levels are significantly altered in human cardiovascular disease as well as in large and small animal cardiovascular disease models. Specifically, β2-spectrin levels were decreased in atrial samples of patients with atrial fibrillation compared with tissue from patients in sinus rhythm. Furthermore, compared with left ventricular samples from nonfailing hearts, β2-spectrin levels were significantly decreased in left ventricle of ischemic- and nonischemic heart failure patients. Left ventricle samples of canine and murine heart failure models confirm reduced β2-spectrin protein levels. Mechanistically, we identify that β2-spectrin levels are tightly regulated by posttranslational mechanisms, namely Ca(2+)- and calpain-dependent proteases. Furthermore, consistent with this data, we observed Ca(2+)- and calpain-dependent loss of β2-spectrin downstream effector proteins, including ankyrin-B in heart. In summary, our findings illustrate that β2-spectrin and downstream molecules are regulated in multiple forms of cardiovascular disease via Ca(2+)- and calpain-dependent proteolysis.
Collapse
Affiliation(s)
- Sakima A Smith
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio;
| | - Langston D Hughes
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Crystal F Kline
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Amber N Kempton
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Lisa E Dorn
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Jerry Curran
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Michael Makara
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Tyler R Webb
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Patrick Wright
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Niels Voigt
- Faculty of Medicine, Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; and
| | - Philip F Binkley
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Paul M L Janssen
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| | - Ahmet Kilic
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Cynthia A Carnes
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Dobromir Dobrev
- Faculty of Medicine, Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; and
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio; Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio; Department of Physiology and Cell Biology, Columbus, Ohio
| |
Collapse
|
23
|
Friedrich O, Reid MB, Van den Berghe G, Vanhorebeek I, Hermans G, Rich MM, Larsson L. The Sick and the Weak: Neuropathies/Myopathies in the Critically Ill. Physiol Rev 2015; 95:1025-109. [PMID: 26133937 PMCID: PMC4491544 DOI: 10.1152/physrev.00028.2014] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Critical illness polyneuropathies (CIP) and myopathies (CIM) are common complications of critical illness. Several weakness syndromes are summarized under the term intensive care unit-acquired weakness (ICUAW). We propose a classification of different ICUAW forms (CIM, CIP, sepsis-induced, steroid-denervation myopathy) and pathophysiological mechanisms from clinical and animal model data. Triggers include sepsis, mechanical ventilation, muscle unloading, steroid treatment, or denervation. Some ICUAW forms require stringent diagnostic features; CIM is marked by membrane hypoexcitability, severe atrophy, preferential myosin loss, ultrastructural alterations, and inadequate autophagy activation while myopathies in pure sepsis do not reproduce marked myosin loss. Reduced membrane excitability results from depolarization and ion channel dysfunction. Mitochondrial dysfunction contributes to energy-dependent processes. Ubiquitin proteasome and calpain activation trigger muscle proteolysis and atrophy while protein synthesis is impaired. Myosin loss is more pronounced than actin loss in CIM. Protein quality control is altered by inadequate autophagy. Ca(2+) dysregulation is present through altered Ca(2+) homeostasis. We highlight clinical hallmarks, trigger factors, and potential mechanisms from human studies and animal models that allow separation of risk factors that may trigger distinct mechanisms contributing to weakness. During critical illness, altered inflammatory (cytokines) and metabolic pathways deteriorate muscle function. ICUAW prevention/treatment is limited, e.g., tight glycemic control, delaying nutrition, and early mobilization. Future challenges include identification of primary/secondary events during the time course of critical illness, the interplay between membrane excitability, bioenergetic failure and differential proteolysis, and finding new therapeutic targets by help of tailored animal models.
Collapse
Affiliation(s)
- O Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - M B Reid
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - G Van den Berghe
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - I Vanhorebeek
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - G Hermans
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - M M Rich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| | - L Larsson
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany; College of Health and Human Performance, University of Florida, Gainesville, Florida; Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; and Department of Physiology and Pharmacology, Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
A Study on Effect of Electroacupuncture on Gene Expression in Hypothalamus of Rats with Stress-Induced Prehypertension Based on Gene Chip Technology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:621237. [PMID: 26229544 PMCID: PMC4502326 DOI: 10.1155/2015/621237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/02/2015] [Indexed: 02/01/2023]
Abstract
Objective. To explore the effect of electroacupuncture (EA) on gene expression in the hypothalamus of rats with stress-induced prehypertension and try to reveal its biological mechanism with gene chip technology. Methods. The stress-induced hypertensive rat model was prepared by combining electric foot-shocks with generated noise. Molding cycle lasted for 14 days and EA intervention was applied on model + EA group during model preparation. Rat Gene 2.0 Array technology was used for the determination of gene expression profiles and the screened key genes were verified by real-time fluorescence quantitative PCR method. Results. Compared with the blank group, 234 genes were upregulated and 73 were downregulated in the model group. Compared with the model group, 110 genes were upregulated and 273 genes were downregulated in model + EA group. The PCR results of the key genes including HSPB1, P2RX4, PPP1R14A, and TH are consistent with that of gene chip test. Conclusion. EA could significantly lower blood pressure of stress-induced prehypertension rats and affect its gene expression profile in hypothalamus. Genes and their signal transduction pathway that related to the contraction of vascular smooth muscle, concentration of Ca(2+), and excitability of sympathetic nerve may be involved in EA's antihypertensive mechanism.
Collapse
|
25
|
Maier LS. Some 'brain' in the heart: a novel microdomain with neuronal Na channels responsible for arrhythmias? Cardiovasc Res 2015; 106:4-5. [PMID: 25681398 DOI: 10.1093/cvr/cvv036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Lars S Maier
- Department for Internal Medicine II, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| |
Collapse
|
26
|
Uryash A, Bassuk J, Kurlansky P, Altamirano F, Lopez JR, Adams JA. Non-invasive technology that improves cardiac function after experimental myocardial infarction: Whole Body Periodic Acceleration (pGz). PLoS One 2015; 10:e0121069. [PMID: 25807532 PMCID: PMC4373845 DOI: 10.1371/journal.pone.0121069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/27/2015] [Indexed: 01/04/2023] Open
Abstract
Myocardial infarction (MI) may produce significant inflammatory changes and adverse ventricular remodeling leading to heart failure and premature death. Pharmacologic, stem cell transplantation, and exercise have not halted the inexorable rise in the prevalence and great economic costs of heart failure despite extensive investigations of such treatments. New therapeutic modalities are needed. Whole Body Periodic Acceleration (pGz) is a non-invasive technology that increases pulsatile shear stress to the endothelium thereby producing several beneficial cardiovascular effects as demonstrated in animal models, normal humans and patients with heart disease. pGz upregulates endothelial derived nitric oxide synthase (eNOS) and its phosphorylation (p-eNOS) to improve myocardial function in models of myocardial stunning and preconditioning. Here we test whether pGz applied chronically after focal myocardial infarction in rats improves functional outcomes from MI. Focal MI was produced by left coronary artery ligation. One day after ligation animals were randomized to receive daily treatments of pGz for four weeks (MI-pGz) or serve as controls (MI-CONT), with an additional group as non-infarction controls (Sham). Echocardiograms and invasive pressure volume loop analysis were carried out. Infarct transmurality, myocardial fibrosis, and markers of inflammatory and anti-inflammatory cytokines were determined along with protein analysis of eNOS, p-eNOS and inducible nitric oxide synthase (iNOS).At four weeks, survival was 80% in MI-pGz vs 50% in MI-CONT (p< 0.01). Ejection fraction and fractional shortening and invasive pressure volume relation indices of afterload and contractility were significantly better in MI-pGz. The latter where associated with decreased infarct transmurality and decreased fibrosis along with increased eNOS, p-eNOS. Additionally, MI-pGz had significantly lower levels of iNOS, inflammatory cytokines (IL-6, TNF-α), and higher level of anti-inflammatory cytokine (IL-10). pGz improved survival and contractile performance, associated with improved myocardial remodeling. pGz may serve as a simple, safe, non-invasive therapeutic modality to improve myocardial function after MI.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| | - Jorge Bassuk
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| | - Paul Kurlansky
- Columbia Heart Source, Columbia University College of Physicians and Surgeons, New York, NY, United States of America
| | - Francisco Altamirano
- Departments of Molecular Bioscience, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Jose R. Lopez
- Departments of Molecular Bioscience, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States of America
| |
Collapse
|
27
|
Balasubramaniam SL, Gopalakrishnapillai A, Gangadharan V, Duncan RL, Barwe SP. Sodium-calcium exchanger 1 regulates epithelial cell migration via calcium-dependent extracellular signal-regulated kinase signaling. J Biol Chem 2015; 290:12463-73. [PMID: 25770213 DOI: 10.1074/jbc.m114.629519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Indexed: 12/16/2022] Open
Abstract
Na(+)/Ca(2+) exchanger-1 (NCX1) is a major calcium extrusion mechanism in renal epithelial cells enabling the efflux of one Ca(2+) ion and the influx of three Na(+) ions. The gradient for this exchange activity is provided by Na,K-ATPase, a hetero-oligomer consisting of a catalytic α-subunit and a regulatory β-subunit (Na,K-β) that also functions as a motility and tumor suppressor. We showed earlier that mice with heart-specific ablation (KO) of Na,K-β had a specific reduction in NCX1 protein and were ouabain-insensitive. Here, we demonstrate that Na,K-β associates with NCX1 and regulates its localization to the cell surface. Madin-Darby canine kidney cells with Na,K-β knockdown have reduced NCX1 protein and function accompanied by 2.1-fold increase in free intracellular calcium and a corresponding increase in the rate of cell migration. Increased intracellular calcium up-regulated ERK1/2 via calmodulin-dependent activation of PI3K. Both myosin light chain kinase and Rho-associated kinase acted as mediators of ERK1/2-dependent migration. Restoring NCX1 expression in β-KD cells reduced migration rate and ERK1/2 activation, suggesting that NCX1 functions downstream of Na,K-β in regulating cell migration. In parallel, inhibition of NCX1 by KB-R7943 in Madin-Darby canine kidney cells, LLC-PK1, and human primary renal epithelial cells (HREpiC) increased ERK1/2 activation and cell migration. This increased migration was associated with high myosin light chain phosphorylation by PI3K/ERK-dependent mechanism in HREpiC cells. These data confirm the role of NCX1 activity in regulating renal epithelial cell migration.
Collapse
Affiliation(s)
- Sona Lakshme Balasubramaniam
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803 and Department of Biological Sciences, University of Delaware, Newark, Delaware 19716
| | - Anilkumar Gopalakrishnapillai
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803 and
| | - Vimal Gangadharan
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716
| | - Randall L Duncan
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716
| | - Sonali P Barwe
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803 and Department of Biological Sciences, University of Delaware, Newark, Delaware 19716
| |
Collapse
|
28
|
Gaitas A, Malhotra R, Li T, Herron T, Jalife J. A device for rapid and quantitative measurement of cardiac myocyte contractility. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2015; 86:034302. [PMID: 25832250 PMCID: PMC4376763 DOI: 10.1063/1.4915500] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/09/2015] [Indexed: 05/27/2023]
Abstract
Cardiac contractility is the hallmark of cardiac function and is a predictor of healthy or diseased cardiac muscle. Despite advancements over the last two decades, the techniques and tools available to cardiovascular scientists are limited in their utility to accurately and reliably measure the amplitude and frequency of cardiomyocyte contractions. Isometric force measurements in the past have entailed cumbersome attachment of isolated and permeabilized cardiomyocytes to a force transducer followed by measurements of sarcomere lengths under conditions of submaximal and maximal Ca(2+) activation. These techniques have the inherent disadvantages of being labor intensive and costly. We have engineered a micro-machined cantilever sensor with an embedded deflection-sensing element that, in preliminary experiments, has demonstrated to reliably measure cardiac cell contractions in real-time. Here, we describe this new bioengineering tool with applicability in the cardiovascular research field to effectively and reliably measure cardiac cell contractility in a quantitative manner. We measured contractility in both primary neonatal rat heart cardiomyocyte monolayers that demonstrated a beat frequency of 3 Hz as well as human embryonic stem cell-derived cardiomyocytes with a contractile frequency of about 1 Hz. We also employed the β-adrenergic agonist isoproterenol (100 nmol l(-1)) and observed that our cantilever demonstrated high sensitivity in detecting subtle changes in both chronotropic and inotropic responses of monolayers. This report describes the utility of our micro-device in both basic cardiovascular research as well as in small molecule drug discovery to monitor cardiac cell contractions.
Collapse
Affiliation(s)
- Angelo Gaitas
- Kytaro, Inc., 11200 SW 8th Street, MARC 430, Miami, Florida 33199, USA
| | - Ricky Malhotra
- Kytaro, Inc., 11200 SW 8th Street, MARC 430, Miami, Florida 33199, USA
| | - Tao Li
- Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Ave., Ann Arbor, Michigan 48109, USA
| | - Todd Herron
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - José Jalife
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
29
|
Koesters A, Engisch KL, Rich MM. Decreased cardiac excitability secondary to reduction of sodium current may be a significant contributor to reduced contractility in a rat model of sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2014; 18:R54. [PMID: 24669759 PMCID: PMC4057164 DOI: 10.1186/cc13800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 03/03/2014] [Indexed: 01/07/2023]
Abstract
Introduction Multisystem organ failure remains a poorly understood complication of sepsis. During sepsis, reduced excitability contributes to organ failure of skeletal muscle, nerves and the spinal cord. The goal of this study was to determine whether reduced excitability might also contribute to cardiac failure during sepsis. Methods Wistar rats were made septic by cecal ligation and puncture. One day later, action potentials were recorded from beating left ventricular papillary muscle ex vivo by impaling myocytes with sharp microelectrodes. Results In cardiac papillary muscle from septic rats, action potential amplitude and rate of rise were reduced, while threshold was elevated. These changes in action potential properties suggest sepsis selectively reduces sodium current. To determine the effects of selective reduction in sodium current, we applied tetrodotoxin to papillary muscle from healthy rats and found reduction in action potential amplitude and rate of rise, as well as elevation of threshold. The changes were similar to those triggered by sepsis. Blocking calcium current using nifedipine did not mimic action potential changes induced by sepsis. Contractility of healthy papillary muscle was reduced to 40% of normal following partial block of sodium current by tetrodotoxin, close to the low contractility of septic papillary muscle, which was 30% of normal. Conclusions Our data suggest cardiac excitability is reduced during sepsis in rats. The reduction in excitability appears to be primarily due to reduction of sodium current. The reduction in sodium current may be sufficient to explain most of the reduction in cardiac contractility during sepsis.
Collapse
|
30
|
Al Kury LT, Yang KHS, Thayyullathil FT, Rajesh M, Ali RM, Shuba YM, Howarth FC, Galadari S, Oz M. Effects of endogenous cannabinoid anandamide on cardiac Na⁺/Ca²⁺ exchanger. Cell Calcium 2014; 55:231-7. [PMID: 24674601 DOI: 10.1016/j.ceca.2014.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/17/2014] [Accepted: 02/23/2014] [Indexed: 10/25/2022]
Abstract
Endocannabinoid anandamide (N-arachidonoyl ethanolamide; AEA) has been shown to cause negative inotropic and antiarrhythmic effects in ventricular myocytes. In this study, using whole-cell patch clamp technique, we have investigated the effects of AEA on cardiac Na(+)/Ca(2+) exchanger (NCX1)-mediated currents. AEA suppressed NCX1 with an IC50 value of 4.7 μM. Both inward and outward components of exchanger currents were suppressed by AEA equally. AEA inhibition was mimicked by the metabolically stable analogue, methanandamide (metAEA, 10 μM) while it was not influenced by inhibition of fatty acid amide hydrolase with 1 μM URB597 incubation. The effect of AEA, was not altered in the presence of cannabinoid receptor 1 and 2 antagonists AM251 (1 μM) and AM630 (1 μM), respectively. In addition, inhibition by AEA remained unchanged after pertussis toxin (PTX, 2 μg/ml) treatment or following the inclusion of GDP-β-S (1 mM) in pipette solution. Currents mediated by NCX1 expressed in HEK-293 cells were also inhibited by 10 μM AEA a partially reversible manner. Confocal microscopy images indicated that the intensity of YFP-NCX1 expression on cell surface was not altered by AEA. Collectively, the results indicate that AEA directly inhibits the function of NCX1 in rat ventricular myocytes and in HEK-293 cells expressing NCX1.
Collapse
Affiliation(s)
- Lina T Al Kury
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Keun-Hang Susan Yang
- Department of Biological Sciences, Schmid College of Science and Engineering, Chapman University, One University Drive, Orange, CA 92866, USA
| | - Faisal T Thayyullathil
- Department of Biochemistry, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Mohanraj Rajesh
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Ramez M Ali
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Yaroslav M Shuba
- Bogomoletz Institute of Physiology and International Center of Molecular Physiology, National Academy of Sciences of Ukraine, Kyiv 24, Ukraine
| | - Frank Christopher Howarth
- Department of Physiology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Sehamuddin Galadari
- Department of Biochemistry, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Murat Oz
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates.
| |
Collapse
|
31
|
Khananshvili D. Sodium-calcium exchangers (NCX): molecular hallmarks underlying the tissue-specific and systemic functions. Pflugers Arch 2013; 466:43-60. [PMID: 24281864 DOI: 10.1007/s00424-013-1405-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/06/2013] [Accepted: 11/09/2013] [Indexed: 12/19/2022]
Abstract
NCX proteins explore the electrochemical gradient of Na(+) to mediate Ca(2+)-fluxes in exchange with Na(+) either in the Ca(2+)-efflux (forward) or Ca(2+)-influx (reverse) mode, whereas the directionality depends on ionic concentrations and membrane potential. Mammalian NCX variants (NCX1-3) and their splice variants are expressed in a tissue-specific manner to modulate the heartbeat rate and contractile force, the brain's long-term potentiation and learning, blood pressure, renal Ca(2+) reabsorption, the immune response, neurotransmitter and insulin secretion, apoptosis and proliferation, mitochondrial bioenergetics, etc. Although the forward mode of NCX represents a major physiological module, a transient reversal of NCX may contribute to EC-coupling, vascular constriction, and synaptic transmission. Notably, the reverse mode of NCX becomes predominant in pathological settings. Since the expression levels of NCX variants are disease-related, the selective pharmacological targeting of tissue-specific NCX variants could be beneficial, thereby representing a challenge. Recent structural and biophysical studies revealed a common module for decoding the Ca(2+)-induced allosteric signal in eukaryotic NCX variants, although the phenotype variances in response to regulatory Ca(2+) remain unclear. The breakthrough discovery of the archaebacterial NCX structure may serve as a template for eukaryotic NCX, although the turnover rates of the transport cycle may differ ~10(3)-fold among NCX variants to fulfill the physiological demands for the Ca(2+) flux rates. Further elucidation of ion-transport and regulatory mechanisms may lead to selective pharmacological targeting of NCX variants under disease conditions.
Collapse
Affiliation(s)
- Daniel Khananshvili
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, Ramat-Aviv, Tel-Aviv, 69978, Israel,
| |
Collapse
|
32
|
Sodium/calcium exchanger is upregulated by sulfide signaling, forms complex with the β1 and β3 but not β2 adrenergic receptors, and induces apoptosis. Pflugers Arch 2013; 466:1329-42. [PMID: 24114174 DOI: 10.1007/s00424-013-1366-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/06/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
Abstract
Hydrogen sulfide (H2S) as a novel gasotransmitter regulates variety of processes, including calcium transport systems. Sodium calcium exchanger (NCX) is one of the key players in a regulation calcium homeostasis. Thus, the aims of our work were to determine effect of sulfide signaling on the NCX type 1 (NCX1) expression and function in HeLa cells, to investigate the relationship of β-adrenergic receptors with the NCX1 in the presence and/or absence of H2S, and to determine physiological importance of this potential communication. As a H2S donor, we used morpholin-4-ium-4-methoxyphenyl(morpholino) phosphinodithioate-GYY4137. We observed increased levels of the NCX1 mRNA, protein, and activity after 24 h of GYY4137 treatment. This increase was accompanied by elevated cAMP due to the GYY4137 treatment, which was completely abolished, when NCX1 was silenced. Increased cAMP levels would point to upregulation of β-adrenergic receptors. Indeed, GYY4137 increased expression of β1 and β3 (but not β2) adrenergic receptors. These receptors co-precipitated, co-localized with the NCX1, and induced apoptosis in the presence of H2S. Our results suggest that sulfide signaling plays a role in regulation of the NCX1, β1 and β3 adrenergic receptors, their co-localization, and stimulation of apoptosis, which might be of a potential importance in cancer treatment.
Collapse
|
33
|
Ottolia M, Torres N, Bridge JHB, Philipson KD, Goldhaber JI. Na/Ca exchange and contraction of the heart. J Mol Cell Cardiol 2013; 61:28-33. [PMID: 23770352 DOI: 10.1016/j.yjmcc.2013.06.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 12/19/2022]
Abstract
Sodium-calcium exchange (NCX) is the major calcium (Ca) efflux mechanism of ventricular cardiomyocytes. Consequently the exchanger plays a critical role in the regulation of cellular Ca content and hence contractility. Reductions in Ca efflux by the exchanger, such as those produced by elevated intracellular sodium (Na) in response to cardiac glycosides, raise sarcoplasmic reticulum (SR) Ca stores. The result is an increased Ca transient and cardiac contractility. Enhanced Ca efflux activity by the exchanger, for example during heart failure, may reduce diadic cleft Ca and excitation-contraction (EC) coupling gain. This aggravates the impaired contractility associated with SR Ca ATPase dysfunction and reduced SR Ca load in failing heart muscle. Recent data from our laboratories indicate that NCX can also impact the efficiency of EC coupling and contractility independent of SR Ca load through diadic cleft priming with Ca during the upstroke of the action potential. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".
Collapse
Affiliation(s)
- Michela Ottolia
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|