1
|
Tashkandi AJ, Gorman A, McGoldrick Mathers E, Carney G, Yacoub A, Setyaningsih WAW, Kuburas R, Margariti A. Metabolic and Mitochondrial Dysregulations in Diabetic Cardiac Complications. Int J Mol Sci 2025; 26:3016. [PMID: 40243689 PMCID: PMC11988959 DOI: 10.3390/ijms26073016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The growing prevalence of diabetes highlights the urgent need to study diabetic cardiovascular complications, specifically diabetic cardiomyopathy, which is a diabetes-induced myocardial dysfunction independent of hypertension or coronary artery disease. This review examines the role of mitochondrial dysfunction in promoting diabetic cardiac dysfunction and highlights metabolic mechanisms such as hyperglycaemia-induced oxidative stress. Chronic hyperglycaemia and insulin resistance can activate harmful pathways, including advanced glycation end-products (AGEs), protein kinase C (PKC) and hexosamine signalling, uncontrolled reactive oxygen species (ROS) production and mishandling of Ca2+ transient. These processes lead to cardiomyocyte apoptosis, fibrosis and contractile dysfunction. Moreover, endoplasmic reticulum (ER) stress and dysregulated RNA-binding proteins (RBPs) and extracellular vesicles (EVs) contribute to tissue damage, which drives cardiac function towards heart failure (HF). Advanced patient-derived induced pluripotent stem cell (iPSC) cardiac organoids (iPS-COs) are transformative tools for modelling diabetic cardiomyopathy and capturing human disease's genetic, epigenetic and metabolic hallmarks. iPS-COs may facilitate the precise examination of molecular pathways and therapeutic interventions. Future research directions encourage the integration of advanced models with mechanistic techniques to promote novel therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Refik Kuburas
- Wellcome Wolfson Institute of Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.J.T.); (A.G.); (E.M.M.); (G.C.); (A.Y.); (W.A.W.S.)
| | - Andriana Margariti
- Wellcome Wolfson Institute of Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.J.T.); (A.G.); (E.M.M.); (G.C.); (A.Y.); (W.A.W.S.)
| |
Collapse
|
2
|
Xue J, Zeng W, John S, Attiq N, Ottolia M, Jiang Y. Structural mechanisms of PIP 2 activation and SEA0400 inhibition in human cardiac sodium-calcium exchanger NCX1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.05.627058. [PMID: 39677781 PMCID: PMC11643123 DOI: 10.1101/2024.12.05.627058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Na+/Ca2+ exchangers (NCXs) transport Ca2+ across the plasma membrane in exchange for Na+ and play a vital role in maintaining cellular Ca2+ homeostasis. Our previous structural study of human cardiac NCX1 (HsNCX1) reveals the overall architecture of the eukaryotic exchanger and the formation of the inactivation assembly by the intracellular regulatory domain that underlies the cytosolic Na+-dependent inactivation and Ca2+ activation of NCX1. Here we present the cryo-EM structures of HsNCX1 in complex with a physiological activator phosphatidylinositol 4,5-bisphosphate (PIP2), or pharmacological inhibitor SEA0400 that enhances the inactivation of the exchanger. We demonstrate that PIP2 binding stimulates NCX1 activity by inducing a conformational change at the interface between the TM and cytosolic domains that destabilizes the inactivation assembly. In contrast, SEA0400 binding in the TM domain of NCX1 stabilizes the exchanger in an inward-facing conformation that facilitates the formation of the inactivation assembly, thereby promoting the Na+-dependent inactivation of NCX1. Thus, this study reveals the structural basis of PIP2 activation and SEA0400 inhibition of NCX1 and provides some mechanistic understandings of cellular regulation and pharmacology of NCX family proteins.
Collapse
Affiliation(s)
- Jing Xue
- Howard Hughes Medical Institute and Department of Physiology, the University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Biophysics, the University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Weizhong Zeng
- Howard Hughes Medical Institute and Department of Physiology, the University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Biophysics, the University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Scott John
- Department of Medicine (Cardiology), UCLA, Los Angeles, California, USA
| | - Nicole Attiq
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - 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
| | - Youxing Jiang
- Howard Hughes Medical Institute and Department of Physiology, the University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Biophysics, the University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
3
|
Piamsiri C, Fefelova N, Pamarthi SH, Gwathmey JK, Chattipakorn SC, Chattipakorn N, Xie LH. Potential Roles of IP 3 Receptors and Calcium in Programmed Cell Death and Implications in Cardiovascular Diseases. Biomolecules 2024; 14:1334. [PMID: 39456267 PMCID: PMC11506173 DOI: 10.3390/biom14101334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a crucial role in maintaining intracellular/cytosolic calcium ion (Ca2+i) homeostasis. The release of Ca2+ from IP3Rs serves as a second messenger and a modulatory factor influencing various intracellular and interorganelle communications during both physiological and pathological processes. Accumulating evidence from in vitro, in vivo, and clinical studies supports the notion that the overactivation of IP3Rs is linked to the pathogenesis of various cardiac conditions. The overactivation of IP3Rs results in the dysregulation of Ca2+ concentration ([Ca2+]) within cytosolic, mitochondrial, and nucleoplasmic cellular compartments. In cardiovascular pathologies, two isoforms of IP3Rs, i.e., IP3R1 and IP3R2, have been identified. Notably, IP3R1 plays a pivotal role in cardiac ischemia and diabetes-induced arrhythmias, while IP3R2 is implicated in sepsis-induced cardiomyopathy and cardiac hypertrophy. Furthermore, IP3Rs have been reported to be involved in various programmed cell death (PCD) pathways, such as apoptosis, pyroptosis, and ferroptosis underscoring their multifaceted roles in cardiac pathophysiology. Based on these findings, it is evident that exploring potential therapeutic avenues becomes crucial. Both genetic ablation and pharmacological intervention using IP3R antagonists have emerged as promising strategies against IP3R-related pathologies suggesting their potential therapeutic potency. This review summarizes the roles of IP3Rs in cardiac physiology and pathology and establishes a foundational understanding with a particular focus on their involvement in the various PCD pathways within the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Chanon Piamsiri
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Sri Harika Pamarthi
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| |
Collapse
|
4
|
Djemai M, Jalouli M, Chahine M. Impacts of DCM-linked gating pore currents on the electrophysiological characteristics of hiPSC-CM monolayers. Biochem Biophys Res Commun 2024; 723:150175. [PMID: 38820625 DOI: 10.1016/j.bbrc.2024.150175] [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: 05/17/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Variants of the SCN5A gene, which encodes the NaV1.5 cardiac sodium channel, have been linked to arrhythmic disorders associated with dilated cardiomyopathy (DCM). However, the precise pathological mechanisms remain elusive. The present study aimed to elucidate the pathophysiological consequences of the DCM-linked Nav1.5/R219H variant, which is known to generate a gating pore current, using patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) cultured in monolayers. METHODS Ventricular- and atrial-like hiPSC-CM monolayers were generated from DCM patients carrying the R219H SCN5A variant as well as from healthy control individuals. CRISPR-corrected hiPSC-CMs served as isogenic controls. Simultaneous optical mapping of action potentials (APs) and calcium transients (CaTs) was employed to measure conduction velocities (CVs) and AP durations (APDs) and served as markers of electrical excitability. Calcium handling was evaluated by assessing CaT uptake (half-time to peak), recapture (tau of decay), and durations (TD50 and TD80). A multi-electrode array (MEA) analysis was conducted on hiPSC-CM monolayers to measure field potential (FP) parameters, including corrected Fridericia FP durations (FPDc). RESULTS Our results revealed that CVs were significantly reduced by more than 50 % in both ventricular- and atrial-like hiPSC-CM monolayers carrying the R219H variant compared to the control group. APDs were also prolonged in the R219H group compared to the control and CRISPR-corrected groups. CaT uptake, reuptake, and duration were also markedly delayed in the R219H group compared to the control and CRISPR-corrected groups in both the ventricular- and the atrial-like hiPSC-CM monolayers. Lastly, the MEA data revealed a notably prolonged FPDc in the ventricular- and atrial-like hiPSC-CMs carrying the R219H variant compared to the control and isogenic control groups. CONCLUSIONS These findings highlight the impact of the gating pore current on AP propagation and calcium homeostasis within a functional syncytium environment and offer valuable insights into the potential mechanisms underlying DCM pathophysiology.
Collapse
Affiliation(s)
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohamed Chahine
- CERVO Brain Research Centre, Quebec City, Quebec, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada.
| |
Collapse
|
5
|
Kong JN, Dipon Ghosh D, Savvidis A, Sando SR, Droste R, Robert Horvitz H. Transcriptional landscape of a hypoxia response identifies cell-specific pathways for adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601765. [PMID: 39005398 PMCID: PMC11245032 DOI: 10.1101/2024.07.02.601765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
How the HIF-1 (Hypoxia-Inducible) transcription factor drives and coordinates distinct responses to low oxygen across diverse cell types is poorly understood. We present a multi-tissue single-cell gene-expression atlas of the hypoxia response of the nematode Caenorhabditis elegans . This atlas highlights how cell-type-specific HIF-1 responses overlap and diverge among and within neuronal, intestinal, and muscle tissues. Using the atlas to guide functional analyses of candidate muscle-specific HIF-1 effectors, we discovered that HIF-1 activation drives downregulation of the tspo-1 ( TSPO, Translocator Protein) gene in vulval muscle cells to modulate a hypoxia-driven change in locomotion caused by contraction of body-wall muscle cells. We further showed that in human cardiomyocytes HIF-1 activation decreases levels of TSPO and thereby alters intracellular cholesterol transport and the mitochondrial network. We suggest that TSPO-1 is an evolutionarily conserved mediator of HIF-1-dependent modulation of muscle and conclude that our gene-expression atlas can help reveal how HIF-1 drives cell-specific adaptations to hypoxia.
Collapse
|
6
|
Scranton K, John S, Angelini M, Steccanella F, Umar S, Zhang R, Goldhaber JI, Olcese R, Ottolia M. Cardiac function is regulated by the sodium-dependent inhibition of the sodium-calcium exchanger NCX1. Nat Commun 2024; 15:3831. [PMID: 38714663 PMCID: PMC11076594 DOI: 10.1038/s41467-024-47850-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/15/2024] [Indexed: 05/10/2024] Open
Abstract
The Na+-Ca2+ exchanger (NCX1) is the dominant Ca2+ extrusion mechanism in cardiac myocytes. NCX1 activity is inhibited by intracellular Na+ via a process known as Na+-dependent inactivation. A central question is whether this inactivation plays a physiological role in heart function. Using CRISPR/Cas9, we inserted the K229Q mutation in the gene (Slc8a1) encoding for NCX1. This mutation removes the Na+-dependent inactivation while preserving transport properties and other allosteric regulations. NCX1 mRNA levels, protein expression, and protein localization are unchanged in K229Q male mice. However, they exhibit reduced left ventricular ejection fraction and fractional shortening, while displaying a prolonged QT interval. K229Q ventricular myocytes show enhanced NCX1 activity, resulting in action potential prolongation, higher incidence of aberrant action potentials, a faster decline of Ca2+ transients, and depressed cell shortening. The results demonstrate that NCX1 Na+-dependent inactivation plays an essential role in heart function by affecting both cardiac excitability and contractility.
Collapse
Affiliation(s)
- Kyle Scranton
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Scott John
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marina Angelini
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Federica Steccanella
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Soban Umar
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rui Zhang
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michela Ottolia
- Department of Anesthesiology & Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Neeman-Egozi S, Livneh I, Dolgopyat I, Nussinovitch U, Milman H, Cohen N, Eisen B, Ciechanover A, Binah O. Stress-Induced Proteasome Sub-Cellular Translocation in Cardiomyocytes Causes Altered Intracellular Calcium Handling and Arrhythmias. Int J Mol Sci 2024; 25:4932. [PMID: 38732146 PMCID: PMC11084437 DOI: 10.3390/ijms25094932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The ubiquitin-proteasome system (UPS) is an essential mechanism responsible for the selective degradation of substrate proteins via their conjugation with ubiquitin. Since cardiomyocytes have very limited self-renewal capacity, as they are prone to protein damage due to constant mechanical and metabolic stress, the UPS has a key role in cardiac physiology and pathophysiology. While altered proteasomal activity contributes to a variety of cardiac pathologies, such as heart failure and ischemia/reperfusion injury (IRI), the environmental cues affecting its activity are still unknown, and they are the focus of this work. Following a recent study by Ciechanover's group showing that amino acid (AA) starvation in cultured cancer cell lines modulates proteasome intracellular localization and activity, we tested two hypotheses in human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs, CMs): (i) AA starvation causes proteasome translocation in CMs, similarly to the observation in cultured cancer cell lines; (ii) manipulation of subcellular proteasomal compartmentalization is associated with electrophysiological abnormalities in the form of arrhythmias, mediated via altered intracellular Ca2+ handling. The major findings are: (i) starving CMs to AAs results in proteasome translocation from the nucleus to the cytoplasm, while supplementation with the aromatic amino acids tyrosine (Y), tryptophan (W) and phenylalanine (F) (YWF) inhibits the proteasome recruitment; (ii) AA-deficient treatments cause arrhythmias; (iii) the arrhythmias observed upon nuclear proteasome sequestration(-AA+YWF) are blocked by KB-R7943, an inhibitor of the reverse mode of the sodium-calcium exchanger NCX; (iv) the retrograde perfusion of isolated rat hearts with AA starvation media is associated with arrhythmias. Collectively, our novel findings describe a newly identified mechanism linking the UPS to arrhythmia generation in CMs and whole hearts.
Collapse
Affiliation(s)
- Shunit Neeman-Egozi
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Ido Livneh
- The Rappaport-Technion Integrated Cancer Center (R-TICC) and The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 319060, Israel; (I.L.); (N.C.)
| | - Irit Dolgopyat
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Udi Nussinovitch
- Department of Cardiology, Edith Wolfson Medical Center, Holon 5822012, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Helena Milman
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Nadav Cohen
- The Rappaport-Technion Integrated Cancer Center (R-TICC) and The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 319060, Israel; (I.L.); (N.C.)
| | - Binyamin Eisen
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Aaron Ciechanover
- The Rappaport-Technion Integrated Cancer Center (R-TICC) and The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 319060, Israel; (I.L.); (N.C.)
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| |
Collapse
|
8
|
Souza Bomfim GH, Mitaishvili E, Schnetkamp PP, Lacruz RS. Na+/Ca2+ exchange in enamel cells is dominated by the K+-dependent NCKX exchanger. J Gen Physiol 2024; 156:e202313372. [PMID: 37947795 PMCID: PMC10637953 DOI: 10.1085/jgp.202313372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/15/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Calcium (Ca2+) extrusion is an essential function of the enamel-forming ameloblasts, providing Ca2+ for extracellular mineralization. The plasma membrane Ca2+ ATPases (PMCAs) remove cytosolic Ca2+ (cCa2+) and were recently shown to be efficient when ameloblasts experienced low cCa2+ elevation. Sodium-calcium (Na+/Ca2+) exchange has higher capacity to extrude cCa2+, but there is limited evidence on the function of the two main families of Na+/Ca2+ exchangers in enamel formation. The purpose of this study was to analyze the function of the NCX (coded by SLC8) and the K+-dependent NCKX (coded by SLC24) exchangers in rat ameloblasts and to compare their efficacy in the two main stages of enamel formation: the enamel forming secretory stage and the mineralizing or maturation stage. mRNA expression profiling confirmed the expression of Slc8 and Slc24 genes in enamel cells, Slc24a4 being the most highly upregulated transcript during the maturation stage, when Ca2+ transport increases. Na+/Ca2+ exchange was analyzed in the Ca2+ influx mode in Fura-2 AM-loaded ameloblasts. We show that maturation-stage ameloblasts have a higher Na+/Ca2+ exchange capacity than secretory-stage cells. We also show that Na+/Ca2+ exchange in both stages is dominated by NCKX over NCX. The importance of NCKX function in ameloblasts may partly explain why mutations in the SLC24A4 gene, but not in SLC8 genes, result in enamel disease. Our results demonstrate that Na+/Ca2+ exchangers are fully operational in ameloblasts and that their contribution to Ca2+ homeostasis increases in the maturation stage, when Ca2+ transport need is higher.
Collapse
Affiliation(s)
| | - Erna Mitaishvili
- Department of Chemistry, Herbert H. Lehman College, City University of New York. PhD Program in Biology, The Graduate Center of The City University of New York, New York, NY, USA
| | - Paul P.M. Schnetkamp
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| |
Collapse
|
9
|
Dattani A, Singh A, McCann GP, Gulsin GS. Myocardial Calcium Handling in Type 2 Diabetes: A Novel Therapeutic Target. J Cardiovasc Dev Dis 2023; 11:12. [PMID: 38248882 PMCID: PMC10817027 DOI: 10.3390/jcdd11010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Type 2 diabetes (T2D) is a multisystem disease with rapidly increasing global prevalence. Heart failure has emerged as a major complication of T2D. Dysregulated myocardial calcium handling is evident in the failing heart and this may be a key driver of cardiomyopathy in T2D, but until recently this has only been demonstrated in animal models. In this review, we describe the physiological concepts behind calcium handling within the cardiomyocyte and the application of novel imaging techniques for the quantification of myocardial calcium uptake. We take an in-depth look at the evidence for the impairment of calcium handling in T2D using pre-clinical models as well as in vivo studies, following which we discuss potential novel therapeutic approaches targeting dysregulated myocardial calcium handling in T2D.
Collapse
Affiliation(s)
- Abhishek Dattani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester LE3 9QP, UK; (A.S.); (G.P.M.); (G.S.G.)
| | | | | | | |
Collapse
|
10
|
Xue J, Zeng W, Han Y, John S, Ottolia M, Jiang Y. Structural mechanisms of the human cardiac sodium-calcium exchanger NCX1. Nat Commun 2023; 14:6181. [PMID: 37794011 PMCID: PMC10550945 DOI: 10.1038/s41467-023-41885-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Na+/Ca2+ exchangers (NCX) transport Ca2+ in or out of cells in exchange for Na+. They are ubiquitously expressed and play an essential role in maintaining cytosolic Ca2+ homeostasis. Although extensively studied, little is known about the global structural arrangement of eukaryotic NCXs and the structural mechanisms underlying their regulation by various cellular cues including cytosolic Na+ and Ca2+. Here we present the cryo-EM structures of human cardiac NCX1 in both inactivated and activated states, elucidating key structural elements important for NCX ion exchange function and its modulation by cytosolic Ca2+ and Na+. We demonstrate that the interactions between the ion-transporting transmembrane (TM) domain and the cytosolic regulatory domain define the activity of NCX. In the inward-facing state with low cytosolic [Ca2+], a TM-associated four-stranded β-hub mediates a tight packing between the TM and cytosolic domains, resulting in the formation of a stable inactivation assembly that blocks the TM movement required for ion exchange function. Ca2+ binding to the cytosolic second Ca2+-binding domain (CBD2) disrupts this inactivation assembly which releases its constraint on the TM domain, yielding an active exchanger. Thus, the current NCX1 structures provide an essential framework for the mechanistic understanding of the ion transport and cellular regulation of NCX family proteins.
Collapse
Affiliation(s)
- Jing Xue
- Howard Hughes Medical Institute and Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weizhong Zeng
- Howard Hughes Medical Institute and Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yan Han
- Howard Hughes Medical Institute and Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Scott John
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Youxing Jiang
- Howard Hughes Medical Institute and Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
Terrar DA. Timing mechanisms to control heart rhythm and initiate arrhythmias: roles for intracellular organelles, signalling pathways and subsarcolemmal Ca 2. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220170. [PMID: 37122228 PMCID: PMC10150226 DOI: 10.1098/rstb.2022.0170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Rhythms of electrical activity in all regions of the heart can be influenced by a variety of intracellular membrane bound organelles. This is true both for normal pacemaker activity and for abnormal rhythms including those caused by early and delayed afterdepolarizations under pathological conditions. The influence of the sarcoplasmic reticulum (SR) on cardiac electrical activity is widely recognized, but other intracellular organelles including lysosomes and mitochondria also contribute. Intracellular organelles can provide a timing mechanism (such as an SR clock driven by cyclic uptake and release of Ca2+, with an important influence of intraluminal Ca2+), and/or can act as a Ca2+ store involved in signalling mechanisms. Ca2+ plays many diverse roles including carrying electric current, driving electrogenic sodium-calcium exchange (NCX) particularly when Ca2+ is extruded across the surface membrane causing depolarization, and activation of enzymes which target organelles and surface membrane proteins. Heart function is also influenced by Ca2+ mobilizing agents (cADP-ribose, nicotinic acid adenine dinucleotide phosphate and inositol trisphosphate) acting on intracellular organelles. Lysosomal Ca2+ release exerts its effects via calcium/calmodulin-dependent protein kinase II to promote SR Ca2+ uptake, and contributes to arrhythmias resulting from excessive beta-adrenoceptor stimulation. A separate arrhythmogenic mechanism involves lysosomes, mitochondria and SR. Interacting intracellular organelles, therefore, have profound effects on heart rhythms and NCX plays a central role. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Derek A Terrar
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
12
|
Bkaily G, Jacques D. Calcium Homeostasis, Transporters, and Blockers in Health and Diseases of the Cardiovascular System. Int J Mol Sci 2023; 24:ijms24108803. [PMID: 37240147 DOI: 10.3390/ijms24108803] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/06/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Calcium is a highly positively charged ionic species. It regulates all cell types' functions and is an important second messenger that controls and triggers several mechanisms, including membrane stabilization, permeability, contraction, secretion, mitosis, intercellular communications, and in the activation of kinases and gene expression. Therefore, controlling calcium transport and its intracellular homeostasis in physiology leads to the healthy functioning of the biological system. However, abnormal extracellular and intracellular calcium homeostasis leads to cardiovascular, skeletal, immune, secretory diseases, and cancer. Therefore, the pharmacological control of calcium influx directly via calcium channels and exchangers and its outflow via calcium pumps and uptake by the ER/SR are crucial in treating calcium transport remodeling in pathology. Here, we mainly focused on selective calcium transporters and blockers in the cardiovascular system.
Collapse
Affiliation(s)
- Ghassan Bkaily
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Danielle Jacques
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
13
|
Ferro LA, Fernandes SLA, Kalinin AL, Monteiro DA. Effects of exposure to sediment-associated fipronil on cardiac function of Neotropical armored catfish Hypostomus regani. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2023; 58:236-245. [PMID: 36803268 DOI: 10.1080/10934529.2023.2182582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Fipronil is widely used as a broad-spectrum insecticide in agriculture, urban environments, and veterinary medicine. Fipronil can enter aquatic ecosystems and spread to sediment and organic matter, representing a risk to non-target species. This study aimed to evaluate the effects of short-term (96 h) exposure to a low and realistic concentration of sediment-associated fipronil (4.2 µg.kg-1 of Regent® 800 WG) on myocardial contractility of armored catfish Hypostomus regain, a benthic fish species. Fipronil exposure induced increased inotropism and acceleration of contractile kinetics, although no alterations in the relative ventricular mass were observed. This better cardiac function was associated with an elevated expression and/or function of the Na+/Ca2+ exchanger and its marked contribution to contraction and relaxation, probably due to a stress-induced adrenergic stimulation. Ventricle strips of exposed fish also exhibited a faster relaxation and a higher cardiac pumping capacity, indicating that armored catfish were able to perform cardiac adjustments to face the exposure. However, a high energetic cost to maintain an increased cardiac performance can make fish more susceptible to other stressors, impairing developmental processes and/or survival. These findings highlight the need for regulations of emerging contaminants, such as fipronil, to ensure adequate protection of the aquatic system.
Collapse
Affiliation(s)
- Lucas Abreu Ferro
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos (UFSCar), São Paulo State University (UNESP), São Carlos, Araraquara, Brazil
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Suzana Luisa Alves Fernandes
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos (UFSCar), São Paulo State University (UNESP), São Carlos, Araraquara, Brazil
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Ana Lúcia Kalinin
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Diana Amaral Monteiro
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| |
Collapse
|
14
|
Valentim M, Brahmbhatt A, Tupling A. Skeletal and cardiac muscle calcium transport regulation in health and disease. Biosci Rep 2022; 42:BSR20211997. [PMID: 36413081 PMCID: PMC9744722 DOI: 10.1042/bsr20211997] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
In healthy muscle, the rapid release of calcium ions (Ca2+) with excitation-contraction (E-C) coupling, results in elevations in Ca2+ concentrations which can exceed 10-fold that of resting values. The sizable transient changes in Ca2+ concentrations are necessary for the activation of signaling pathways, which rely on Ca2+ as a second messenger, including those involved with force generation, fiber type distribution and hypertrophy. However, prolonged elevations in intracellular Ca2+ can result in the unwanted activation of Ca2+ signaling pathways that cause muscle damage, dysfunction, and disease. Muscle employs several calcium handling and calcium transport proteins that function to rapidly return Ca2+ concentrations back to resting levels following contraction. This review will detail our current understanding of calcium handling during the decay phase of intracellular calcium transients in healthy skeletal and cardiac muscle. We will also discuss how impairments in Ca2+ transport can occur and how mishandling of Ca2+ can lead to the pathogenesis and/or progression of skeletal muscle myopathies and cardiomyopathies.
Collapse
Affiliation(s)
- Mark A. Valentim
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - Aditya N. Brahmbhatt
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - A. Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| |
Collapse
|
15
|
Lim SY, Lim FLS, Criado-Navarro I, Yeo XH, Dayal H, Vemulapalli SD, Seah SJ, Laserna AKC, Yang X, Tan SH, Chan MY, Li SFY. Multi-Omics Investigation into Acute Myocardial Infarction: An Integrative Method Revealing Interconnections amongst the Metabolome, Lipidome, Glycome, and Metallome. Metabolites 2022; 12:1080. [PMID: 36355163 PMCID: PMC9693522 DOI: 10.3390/metabo12111080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Acute myocardial infarction (AMI) is a leading cause of mortality and morbidity worldwide. This work aims to investigate the translational potential of a multi-omics study (comprising metabolomics, lipidomics, glycomics, and metallomics) in revealing biomechanistic insights into AMI. Following the N-glycomics and metallomics studies performed by our group previously, untargeted metabolomic and lipidomic profiles were generated and analysed in this work via the use of a simultaneous metabolite/lipid extraction and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis workflow. The workflow was applied to blood plasma samples from AMI cases (n = 101) and age-matched healthy controls (n = 66). The annotated metabolomic (number of features, n = 27) and lipidomic (n = 48) profiles, along with the glycomic (n = 37) and metallomic (n = 30) profiles of the same set of AMI and healthy samples were integrated and analysed. The integration method used here works by identifying a linear combination of maximally correlated features across the four omics datasets, via utilising both block-partial least squares-discriminant analysis (block-PLS-DA) based on sparse generalised canonical correlation analysis. Based on the multi-omics mapping of biomolecular interconnections, several postulations were derived. These include the potential roles of glycerophospholipids in N-glycan-modulated immunoregulatory effects, as well as the augmentation of the importance of Ca-ATPases in cardiovascular conditions, while also suggesting contributions of phosphatidylethanolamine in their functions. Moreover, it was shown that combining the four omics datasets synergistically enhanced the classifier performance in discriminating between AMI and healthy subjects. Fresh and intriguing insights into AMI, otherwise undetected via single-omics analysis, were revealed in this multi-omics study. Taken together, we provide evidence that a multi-omics strategy may synergistically reinforce and enhance our understanding of diseases.
Collapse
Affiliation(s)
- Si Ying Lim
- NUS Graduate School’s Integrative Sciences & Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Felicia Li Shea Lim
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | | | - Xin Hao Yeo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Hiranya Dayal
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | | | - Song Jie Seah
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Anna Karen Carrasco Laserna
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- Central Instrumentation Facility (Laguna Campus), Office of the Vice President for Research and Innovation, De La Salle University, Manila 1004, Philippines
| | - Xiaoxun Yang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Sock Hwee Tan
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Mark Y. Chan
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Sam Fong Yau Li
- NUS Graduate School’s Integrative Sciences & Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
16
|
Macías Á, González-Guerra A, Moreno-Manuel AI, Cruz FM, Gutiérrez LK, García-Quintáns N, Roche-Molina M, Bermúdez-Jiménez F, Andrés V, Vera-Pedrosa ML, Martínez-Carrascoso I, Bernal JA, Jalife J. Kir2.1 dysfunction at the sarcolemma and the sarcoplasmic reticulum causes arrhythmias in a mouse model of Andersen-Tawil syndrome type 1. NATURE CARDIOVASCULAR RESEARCH 2022; 1:900-917. [PMID: 39195979 PMCID: PMC11358039 DOI: 10.1038/s44161-022-00145-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/02/2022] [Indexed: 08/29/2024]
Abstract
Andersen-Tawil syndrome type 1 (ATS1) is associated with life-threatening arrhythmias of unknown mechanism. In this study, we generated and characterized a mouse model of ATS1 carrying the trafficking-deficient mutant Kir2.1Δ314-315 channel. The mutant mouse recapitulates the electrophysiological phenotype of ATS1, with QT prolongation exacerbated by flecainide or isoproterenol, drug-induced QRS prolongation, increased vulnerability to reentrant arrhythmias and multifocal discharges resembling catecholaminergic polymorphic ventricular tachycardia (CPVT). Kir2.1Δ314-315 cardiomyocytes display significantly reduced inward rectifier K+ and Na+ currents, depolarized resting membrane potential and prolonged action potentials. We show that, in wild-type mouse cardiomyocytes and skeletal muscle cells, Kir2.1 channels localize to sarcoplasmic reticulum (SR) microdomains, contributing to intracellular Ca2+ homeostasis. Kir2.1Δ314-315 cardiomyocytes exhibit defective SR Kir2.1 localization and function, as intact and permeabilized Kir2.1Δ314-315 cardiomyocytes display abnormal spontaneous Ca2+ release events. Overall, defective Kir2.1 channel function at the sarcolemma and the SR explain the life-threatening arrhythmias in ATS1 and its overlap with CPVT.
Collapse
Affiliation(s)
- Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lilian K Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Marta Roche-Molina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | | | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
17
|
Hegner P, Drzymalski M, Biedermann A, Memmel B, Durczok M, Wester M, Floerchinger B, Provaznik Z, Schmid C, Zausig Y, Maier LS, Wagner S. SAR296968, a Novel Selective Na+/Ca2+ Exchanger Inhibitor, Improves Ca2+ Handling and Contractile Function in Human Atrial Cardiomyocytes. Biomedicines 2022; 10:biomedicines10081932. [PMID: 36009478 PMCID: PMC9406204 DOI: 10.3390/biomedicines10081932] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/30/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background: In reverse-mode, cardiac sodium-calcium exchanger (NCX) can increase the cytoplasmic Ca2+ concentration in response to high intracellular Na+ levels, which may contribute to diastolic contractile dysfunction. Furthermore, increased spontaneous Ca2+ release from intracellular stores can activate forward mode NCX. The resulting transient inward current causes delayed afterdepolarization (DAD)-dependent arrhythmias. Moreover, recently, NCX has been associated with impaired relaxation and reduced cardiac function in heart failure with preserved ejection fraction (HFpEF). Since NCX is upregulated in human chronic atrial fibrillation (AF) as well as heart failure (HF), specific inhibition may have therapeutic potential. Objective: We tested the antiarrhythmic, lusitropic and inotropic effects of a novel selective NCX-inhibitor (SAR296968) in human atrial myocardium. Methods and Results: Right atrial appendage biopsies of 46 patients undergoing elective cardiac surgery in a predominant HFpEF cohort (n = 24/46) were investigated. In isolated human atrial cardiomyocytes, SAR296968 reduced the frequency of spontaneous SR Ca2+ release events and increased caffeine transient amplitude. In accordance, in isolated atrial trabeculae, SAR296968 enhanced the developed tension after a 30 s pause of electrical stimulation consistent with reduced diastolic sarcoplasmic reticulum (SR) Ca2+ leak. Moreover, compared to vehicle, SAR296968 decreased steady-state diastolic tension (at 1 Hz) without impairing developed systolic tension. Importantly, SAR296968 did not affect the safety parameters, such as resting membrane potential or action potential duration as measured by patch clamp. Conclusion: The novel selective NCX-inhibitor SAR296968 inhibits atrial pro-arrhythmic activity and improves diastolic and contractile function in human atrial myocardium, which may have therapeutic implications, especially for treatment of HFpEF.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Marzena Drzymalski
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Alexander Biedermann
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Bernadette Memmel
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Melanie Durczok
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Michael Wester
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Bernhard Floerchinger
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Zdenek Provaznik
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - York Zausig
- Department of Anesthesiology, University Medical Center Regensburg, 93053 Regensburg, Germany
- Department of Anesthesiology and Operative Intensive Care Medicine, Aschaffenburg-Alzenau Hospital, 63739 Aschaffenburg, Germany
| | - Lars S. Maier
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-944-7206
| |
Collapse
|
18
|
Lopina OD, Fedorov DA, Sidorenko SV, Bukach OV, Klimanova EA. Sodium Ions as Regulators of Transcription in Mammalian Cells. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:789-799. [PMID: 36171659 DOI: 10.1134/s0006297922080107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 06/16/2023]
Abstract
The maintenance of an uneven distribution of Na+ and K+ ions between the cytoplasm and extracellular medium is the basis for the functioning of any animal cell. Changes in the intracellular ratio of these cations occur in response to numerous stimuli and are important for the cell activity regulation. Numerous experimental data have shown that gene transcription in mammalian cells can be regulated by changes in the intracellular [Na+]i/[K+]i ratio. Here, we discuss possible mechanisms of such regulation in various cell types, with special attention to the [Ca2+]-independent signaling pathways that suggest the presence of an intracellular sensor of monovalent cations. As such sensor, we propose the secondary structures of nucleic acids called G-quadruplexes. They are widely represented in mammalian genomes and are often found in the promoters of genes encoding transcription factors.
Collapse
Affiliation(s)
- Olga D Lopina
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia.
| | - Dmitrii A Fedorov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | | | - Olesya V Bukach
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | | |
Collapse
|
19
|
Rodrigues T, Piccirillo S, Magi S, Preziuso A, Dos Santos Ramos V, Serfilippi T, Orciani M, Maciel Palacio Alvarez M, Luis Dos Santos Tersariol I, Amoroso S, Lariccia V. Control of Ca 2+ and metabolic homeostasis by the Na +/Ca 2+ exchangers (NCXs) in health and disease. Biochem Pharmacol 2022; 203:115163. [PMID: 35803319 DOI: 10.1016/j.bcp.2022.115163] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022]
Abstract
Spatial and temporal control of calcium (Ca2+) levels is essential for the background rhythms and responses of living cells to environmental stimuli. Whatever other regulators a given cellular activity may have, localized and wider scale Ca2+ events (sparks, transients, and waves) are hierarchical determinants of fundamental processes such as cell contraction, excitability, growth, metabolism and survival. Different cell types express specific channels, pumps and exchangers to efficiently generate and adapt Ca2+ patterns to cell requirements. The Na+/Ca2+ exchangers (NCXs) in particular contribute to Ca2+ homeostasis by buffering intracellular Ca2+ loads according to the electrochemical gradients of substrate ions - i.e., Ca2+ and sodium (Na+) - and under a dynamic control of redundant regulatory processes. An interesting feature of NCX emerges from the strict relationship that connects transporter activity with cell metabolism: on the one hand NCX operates under constant control of ATP-dependent regulatory processes, on the other hand the ion fluxes generated through NCX provide mechanistic support for the Na+-driven uptake of glutamate and Ca2+ influx to fuel mitochondrial respiration. Proof of concept evidence highlights therapeutic potential of preserving a timed and balanced NCX activity in a growing rate of diseases (including excitability, neurodegenerative, and proliferative disorders) because of an improved ability of stressed cells to safely maintain ion gradients and mitochondrial bioenergetics. Here, we will summarize and review recent works that have focused on the pathophysiological roles of NCXs in balancing the two-way relationship between Ca2+ signals and metabolism.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vyctória Dos Santos Ramos
- Interdisciplinary Center for Biochemistry Investigation (CIIB), University of Mogi das Cruzes (UMC), Mogi das Cruzes, SP, Brazil
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Histology, University "Politecnica delle Marche", Ancona, Italy.
| | - Marcela Maciel Palacio Alvarez
- Department of Biochemistry, São Paulo School of Medicine, Federal University of São Paulo (Unifesp) São Paulo, SP, Brazil
| | | | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| |
Collapse
|
20
|
Song L, Bekdash R, Morikawa K, Quejada JR, Klein AD, Aina-Badejo D, Yoshida K, Yamamoto HE, Chalan A, Yang R, Patel A, Sirabella D, Lee TM, Joseph LC, Kawano F, Warren JS, Soni RK, Morrow JP, Yazawa M. Sigma non-opioid receptor 1 is a potential therapeutic target for long QT syndrome. NATURE CARDIOVASCULAR RESEARCH 2022; 1:142-156. [PMID: 36051854 PMCID: PMC9431959 DOI: 10.1038/s44161-021-00016-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Some missense gain-of-function mutations in CACNA1C gene, encoding calcium channel CaV1.2, cause a life-threatening form of long QT syndrome named Timothy syndrome, with currently no clinically-effective therapeutics. Here we report that pharmacological targeting of sigma non-opioid intracellular receptor 1 (SIGMAR1) can restore electrophysiological function in iPSC-derived cardiomyocytes generated from patients with Timothy syndrome and two common forms of long QT syndrome, type 1 (LQTS1) and 2 (LQTS2), caused by missense trafficking mutations in potassium channels. Electrophysiological recordings demonstrate that an FDA-approved cough suppressant, dextromethorphan, can be used as an agonist of SIGMAR1, to shorten the prolonged action potential in Timothy syndrome cardiomyocytes and human cellular models of LQTS1 and LQTS2. When tested in vivo, dextromethorphan also normalized the prolonged QT intervals in Timothy syndrome model mice. Overall, our study demonstrates that SIGMAR1 is a potential therapeutic target for Timothy syndrome and possibly other inherited arrhythmias such as LQTS1 and LQTS2.
Collapse
|
21
|
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
|
22
|
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: 23] [Impact Index Per Article: 5.8] [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
|
23
|
Pan-phylum genome-wide identification of sodium calcium exchangers reveal heterogeneous expansions and possible roles in nematode parasitism. Gene 2021; 810:146052. [PMID: 34756961 DOI: 10.1016/j.gene.2021.146052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
Calcium signaling is ubiquitous in nematode development from fertilization to cell specification to apoptosis. Calcium also regulates dauer entry in Caenorhabditis elegans, which corresponds to the infective stage of parasitic nematodes. In diverse parasites such as Trypanosoma cruzi and Toxoplasma gondii calcium has been shown to regulate host cell entry and egress, and perturbing calcium signaling represents a possible route to inhibit infection and parasitism in these species. Sodium calcium exchangers are considered the most important mechanism of calcium efflux, and our lab has previously characterized the sodium calcium exchanger gene family in C. elegans and studied the diversity of this family across a subset of specific nematode species. Here we build upon these data and explore sodium calcium exchangers across 108 species of nematodes. Our data reveal substantial differences in sodium calcium exchanger counts across the Phylum and detail expansions and contractions of specific exchanger subtypes within certain nematode clades. Finally, we also provide evidence for a role of sodium calcium exchangers in parasite activation by examining differentially expressed genes in non-activated versus activated infective stage larvae. Taken together our findings paint a heterogeneous picture of sodium calcium exchanger evolution across the Phylum Nematoda that may reflect unique adaptations to free-living and parasitic lifestyles.
Collapse
|
24
|
Fischesser DM, Bo B, Benton RP, Su H, Jahanpanah N, Haworth KJ. Controlling Reperfusion Injury With Controlled Reperfusion: Historical Perspectives and New Paradigms. J Cardiovasc Pharmacol Ther 2021; 26:504-523. [PMID: 34534022 DOI: 10.1177/10742484211046674] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac reperfusion injury is a well-established outcome following treatment of acute myocardial infarction and other types of ischemic heart conditions. Numerous cardioprotection protocols and therapies have been pursued with success in pre-clinical models. Unfortunately, there has been lack of successful large-scale clinical translation, perhaps in part due to the multiple pathways that reperfusion can contribute to cell death. The search continues for new cardioprotection protocols based on what has been learned from past results. One class of cardioprotection protocols that remain under active investigation is that of controlled reperfusion. This class consists of those approaches that modify, in a controlled manner, the content of the reperfusate or the mechanical properties of the reperfusate (e.g., pressure and flow). This review article first provides a basic overview of the primary pathways to cell death that have the potential to be addressed by various forms of controlled reperfusion, including no-reflow phenomenon, ion imbalances (particularly calcium overload), and oxidative stress. Descriptions of various controlled reperfusion approaches are described, along with summaries of both mechanistic and outcome-oriented studies at the pre-clinical and clinical phases. This review will constrain itself to approaches that modify endogenously-occurring blood components. These approaches include ischemic postconditioning, gentle reperfusion, controlled hypoxic reperfusion, controlled hyperoxic reperfusion, controlled acidotic reperfusion, and controlled ionic reperfusion. This review concludes with a discussion of the limitations of past approaches and how they point to potential directions of investigation for the future.
Collapse
Affiliation(s)
- Demetria M Fischesser
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Bin Bo
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Rachel P Benton
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Haili Su
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Newsha Jahanpanah
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| | - Kevin J Haworth
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, College of Medicine, 2514University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
25
|
Weissman D, Maack C. Redox signaling in heart failure and therapeutic implications. Free Radic Biol Med 2021; 171:345-364. [PMID: 34019933 DOI: 10.1016/j.freeradbiomed.2021.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/17/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Heart failure is a growing health burden worldwide characterized by alterations in excitation-contraction coupling, cardiac energetic deficit and oxidative stress. While current treatments are mostly limited to antagonization of neuroendocrine activation, more recent data suggest that also targeting metabolism may provide substantial prognostic benefit. However, although in a broad spectrum of preclinical models, oxidative stress plays a causal role for the development and progression of heart failure, no treatment that targets reactive oxygen species (ROS) directly has entered the clinical arena yet. In the heart, ROS derive from various sources, such as NADPH oxidases, xanthine oxidase, uncoupled nitric oxide synthase and mitochondria. While mitochondria are the primary source of ROS in the heart, communication between different ROS sources may be relevant for physiological signalling events as well as pathologically elevated ROS that deteriorate excitation-contraction coupling, induce hypertrophy and/or trigger cell death. Here, we review the sources of ROS in the heart, the modes of pathological activation of ROS formation as well as therapeutic approaches that may target ROS specifically in mitochondria.
Collapse
Affiliation(s)
- David Weissman
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany; Department of Internal Medicine 1, University Clinic Würzburg, Würzburg, Germany.
| |
Collapse
|
26
|
Shemer Y, Mekies LN, Ben Jehuda R, Baskin P, Shulman R, Eisen B, Regev D, Arbustini E, Gerull B, Gherghiceanu M, Gottlieb E, Arad M, Binah O. Investigating LMNA-Related Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22157874. [PMID: 34360639 PMCID: PMC8346174 DOI: 10.3390/ijms22157874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/09/2023] Open
Abstract
LMNA-related dilated cardiomyopathy is an inherited heart disease caused by mutations in the LMNA gene encoding for lamin A/C. The disease is characterized by left ventricular enlargement and impaired systolic function associated with conduction defects and ventricular arrhythmias. We hypothesized that LMNA-mutated patients' induced Pluripotent Stem Cell-derived cardiomyocytes (iPSC-CMs) display electrophysiological abnormalities, thus constituting a suitable tool for deciphering the arrhythmogenic mechanisms of the disease, and possibly for developing novel therapeutic modalities. iPSC-CMs were generated from two related patients (father and son) carrying the same E342K mutation in the LMNA gene. Compared to control iPSC-CMs, LMNA-mutated iPSC-CMs exhibited the following electrophysiological abnormalities: (1) decreased spontaneous action potential beat rate and decreased pacemaker current (If) density; (2) prolonged action potential duration and increased L-type Ca2+ current (ICa,L) density; (3) delayed afterdepolarizations (DADs), arrhythmias and increased beat rate variability; (4) DADs, arrhythmias and cessation of spontaneous firing in response to β-adrenergic stimulation and rapid pacing. Additionally, compared to healthy control, LMNA-mutated iPSC-CMs displayed nuclear morphological irregularities and gene expression alterations. Notably, KB-R7943, a selective inhibitor of the reverse-mode of the Na+/Ca2+ exchanger, blocked the DADs in LMNA-mutated iPSC-CMs. Our findings demonstrate cellular electrophysiological mechanisms underlying the arrhythmias in LMNA-related dilated cardiomyopathy.
Collapse
Affiliation(s)
- Yuval Shemer
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
| | - Lucy N. Mekies
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
| | - Ronen Ben Jehuda
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
- Department of Biotechnology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| | - Polina Baskin
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
| | - Rita Shulman
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
| | - Binyamin Eisen
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
| | - Danielle Regev
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
| | - Eloisa Arbustini
- Centre for Inherited Cardiovascular Diseases, IRCCS Foundation, Policlinico San Matteo, 27100 Pavia, Italy;
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany;
| | | | - Eyal Gottlieb
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 31096, Israel;
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Rappaport Research Institute, Technion—Israel Institute of Technology, Haifa 31096, Israel; (Y.S.); (L.N.M.); (R.B.J.); (P.B.); (R.S.); (B.E.); (D.R.)
- Correspondence: ; Tel.: +972-4-8295262; Fax: +972-4-8513919
| |
Collapse
|
27
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
28
|
Goerg J, Sommerfeld M, Greiner B, Lauer D, Seckin Y, Kulikov A, Ivkin D, Kintscher U, Okovityi S, Kaschina E. Low-Dose Empagliflozin Improves Systolic Heart Function after Myocardial Infarction in Rats: Regulation of MMP9, NHE1, and SERCA2a. Int J Mol Sci 2021; 22:ijms22115437. [PMID: 34063987 PMCID: PMC8196699 DOI: 10.3390/ijms22115437] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022] Open
Abstract
The effects of the selective sodium-glucose cotransporter 2 (SGLT2) inhibitor empagliflozin in low dose on cardiac function were investigated in normoglycemic rats. Cardiac parameters were measured by intracardiac catheterization 30 min after intravenous application of empagliflozin to healthy animals. Empagliflozin increased the ventricular systolic pressure, mean pressure, and the max dP/dt (p < 0.05). Similarly, treatment with empagliflozin (1 mg/kg, p.o.) for one week increased the cardiac output, stroke volume, and fractional shortening (p < 0.05). Myocardial infarction (MI) was induced by ligation of the left coronary artery. On day 7 post MI, empagliflozin (1 mg/kg, p.o.) improved the systolic heart function as shown by the global longitudinal strain (−21.0 ± 1.1% vs. −16.6 ± 0.7% in vehicle; p < 0.05). In peri-infarct tissues, empagliflozin decreased the protein expression of matrix metalloproteinase 9 (MMP9) and favorably regulated the cardiac transporters sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) and sodium hydrogen exchanger 1 (NHE1). In H9c2 cardiac cells, empagliflozin decreased the MMP2,9 activity and prevented apoptosis. Empagliflozin did not alter the arterial stiffness, blood pressure, markers of fibrosis, and necroptosis. Altogether, short-term treatment with low-dose empagliflozin increased the cardiac contractility in normoglycemic rats and improved the systolic heart function in the early phase after MI. These effects are attributed to a down-regulation of MMP9 and NHE1, and an up-regulation of SERCA2a. This study is of clinical importance because it suggests that a low-dose treatment option with empagliflozin may improve cardiovascular outcomes post-MI. Down-regulation of MMPs could be relevant to many remodeling processes including cancer disease.
Collapse
Affiliation(s)
- Jana Goerg
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
| | - Manuela Sommerfeld
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
| | - Bettina Greiner
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
| | - Dilyara Lauer
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
| | - Yasemin Seckin
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
- Department of Biotechnology, University of Applied Science, 13353 Berlin, Germany
| | - Alexander Kulikov
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia;
| | - Dmitry Ivkin
- Saint-Petersburg State Chemical-Pharmaceutical University, 197376 Saint Petersburg, Russia; (D.I.); (S.O.)
| | - Ulrich Kintscher
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
| | - Sergey Okovityi
- Saint-Petersburg State Chemical-Pharmaceutical University, 197376 Saint Petersburg, Russia; (D.I.); (S.O.)
| | - Elena Kaschina
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Center for Cardiovascular Research (CCR), 10115 Berlin, Germany; (J.G.); (M.S.); (B.G.); (D.L.); (Y.S.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-525-024
| |
Collapse
|
29
|
Gök C, Plain F, Robertson AD, Howie J, Baillie GS, Fraser NJ, Fuller W. Dynamic Palmitoylation of the Sodium-Calcium Exchanger Modulates Its Structure, Affinity for Lipid-Ordered Domains, and Inhibition by XIP. Cell Rep 2021; 31:107697. [PMID: 32521252 PMCID: PMC7296346 DOI: 10.1016/j.celrep.2020.107697] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/07/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
The transmembrane sodium-calcium (Na-Ca) exchanger 1 (NCX1) regulates cytoplasmic Ca levels by facilitating electrogenic exchange of Ca for Na. Palmitoylation, the only reversible post-translational modification known to modulate NCX1 activity, controls NCX1 inactivation. Here, we show that palmitoylation of NCX1 modifies the structural arrangement of the NCX1 dimer and controls its affinity for lipid-ordered membrane domains. NCX1 palmitoylation occurs dynamically at the cell surface under the control of the enzymes zDHHC5 and APT1. We identify the position of the endogenous exchange inhibitory peptide (XIP) binding site within the NCX1 regulatory intracellular loop and demonstrate that palmitoylation controls the ability of XIP to bind this site. We also show that changes in NCX1 palmitoylation change cytosolic Ca. Our results thus demonstrate the broad molecular consequences of NCX1 palmitoylation and highlight a means to manipulate the inactivation of this ubiquitous ion transporter that could ameliorate pathologies linked to Ca overload via NCX1. NCX1 is dynamically palmitoylated at the cell surface by zDHHC5 and APT1 Palmitoylation modifies the NCX1 dimer’s structure and affinity for lipid rafts We identify the binding site of the endogenous XIP domain in NCX1’s regulatory loop Palmitoylation modifies NCX1 XIP affinity and hence regulates intracellular Ca
Collapse
Affiliation(s)
- Caglar Gök
- Institute of Cardiovascular & Medical Sciences, Sir James Black Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - Fiona Plain
- School of Medicine, Ninewells Hospital, University of Dundee, Dundee DD1 9SY, UK
| | - Alan D Robertson
- Institute of Cardiovascular & Medical Sciences, Sir James Black Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - Jacqueline Howie
- Institute of Cardiovascular & Medical Sciences, Sir James Black Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - George S Baillie
- Institute of Cardiovascular & Medical Sciences, Sir James Black Building, University of Glasgow, Glasgow G12 8QQ, UK
| | - Niall J Fraser
- School of Medicine, Ninewells Hospital, University of Dundee, Dundee DD1 9SY, UK
| | - William Fuller
- Institute of Cardiovascular & Medical Sciences, Sir James Black Building, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
30
|
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] [MESH Headings] [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
|
31
|
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] [MESH Headings] [Grants] [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. SIGNIFICANCE STATEMENT: Ethanol acutely inhibits hippocampal NMDARs, LTP, and learning. This study found that certain oxysterols that are NMDAR-positive allosteric modulators can overcome the acute effects of ethanol on NMDARs, LTP, and learning. Oxysterols differ in their effects from agents that inhibit integrated cellular stress responses.
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
|
32
|
Zhao R, Liu X, Qi Z, Yao X, Tsang SY. TRPV1 channels regulate the automaticity of embryonic stem cell-derived cardiomyocytes through stimulating the Na + /Ca 2+ exchanger current. J Cell Physiol 2021; 236:6806-6823. [PMID: 33782967 DOI: 10.1002/jcp.30369] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 02/25/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022]
Abstract
Calcium controls the excitation-contraction coupling in cardiomyocytes. Embryonic stem cell-derived cardiomyocytes (ESC-CMs) are an important cardiomyocyte source for regenerative medicine and drug screening. Transient receptor potential vanilloid 1 (TRPV1) channels are nonselective cation channels that permeate sodium and calcium. This study aimed to investigate whether TRPV1 channels regulate the electrophysiological characteristics of ESC-CMs. If yes, what is the mechanism behind? By immunostaining and subcellular fractionation, followed by western blotting, TRPV1 was found to locate intracellularly. The staining pattern of TRPV1 was found to largely overlap with that of the sarco/endoplasmic reticulum Ca2+ -ATPase, the sarcoplasmic reticulum (SR) marker. By electrophysiology and calcium imaging, pharmacological blocker of TRPV1 and the molecular tool TRPV1β (which could functionally knockdown TRPV1) were found to decrease the rate and diastolic depolarization slope of spontaneous action potentials, and the amplitude and frequency of global calcium transients. By calcium imaging, in the absence of external calcium, TRPV1-specific opener increased intracellular calcium; this increase was abolished by preincubation with caffeine, which could deplete SR calcium store. The results suggest that TRPV1 controls calcium release from the SR. By electrophysiology, TRPV1 blockade and functional knockdown of TRPV1 decreased the Na+ /Ca2+ exchanger (NCX) currents from both the forward and reverse modes, suggesting that sodium and calcium through TRPV1 stimulate the NCX activity. Our novel findings suggest that TRPV1 activity is important for regulating the spontaneous activity of ESC-CMs and reveal a novel interplay between TRPV1 and NCX in regulating the physiological functions of ESC-CMs.
Collapse
Affiliation(s)
- Rui Zhao
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xianji Liu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zenghua Qi
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Suk Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
33
|
Rufaihah AJ, Chen CK, Yap CH, Mattar CNZ. Mending a broken heart: In vitro, in vivo and in silico models of congenital heart disease. Dis Model Mech 2021; 14:dmm047522. [PMID: 33787508 PMCID: PMC8033415 DOI: 10.1242/dmm.047522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Birth defects contribute to ∼0.3% of global infant mortality in the first month of life, and congenital heart disease (CHD) is the most common birth defect among newborns worldwide. Despite the significant impact on human health, most treatments available for this heterogenous group of disorders are palliative at best. For this reason, the complex process of cardiogenesis, governed by multiple interlinked and dose-dependent pathways, is well investigated. Tissue, animal and, more recently, computerized models of the developing heart have facilitated important discoveries that are helping us to understand the genetic, epigenetic and mechanobiological contributors to CHD aetiology. In this Review, we discuss the strengths and limitations of different models of normal and abnormal cardiogenesis, ranging from single-cell systems and 3D cardiac organoids, to small and large animals and organ-level computational models. These investigative tools have revealed a diversity of pathogenic mechanisms that contribute to CHD, including genetic pathways, epigenetic regulators and shear wall stresses, paving the way for new strategies for screening and non-surgical treatment of CHD. As we discuss in this Review, one of the most-valuable advances in recent years has been the creation of highly personalized platforms with which to study individual diseases in clinically relevant settings.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Ching Kit Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Choon Hwai Yap
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat -National University Children's Medical Institute, National University Health System, Singapore 119228
- Department of Bioengineering, Imperial College London, London, UK
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore 119228
| |
Collapse
|
34
|
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
|
35
|
Stress-driven cardiac calcium mishandling via a kinase-to-kinase crosstalk. Pflugers Arch 2021; 473:363-375. [PMID: 33590296 PMCID: PMC7940337 DOI: 10.1007/s00424-021-02533-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 01/25/2023]
Abstract
Calcium homeostasis in the cardiomyocyte is critical to the regulation of normal cardiac function. Abnormal calcium dynamics such as altered uptake by the sarcoplasmic reticulum (SR) Ca2+-ATPase and increased diastolic SR calcium leak are involved in the development of maladaptive cardiac remodeling under pathological conditions. Ca2+/calmodulin-dependent protein kinase II-δ (CaMKIIδ) is a well-recognized key molecule in calcium dysregulation in cardiomyocytes. Elevated cellular stress is known as a common feature during pathological remodeling, and c-jun N-terminal kinase (JNK) is an important stress kinase that is activated in response to intrinsic and extrinsic stress stimuli. Our lab recently identified specific actions of JNK isoform 2 (JNK2) in CaMKIIδ expression, activation, and CaMKIIδ-dependent SR Ca2+ mishandling in the stressed heart. This review focuses on the current understanding of cardiac SR calcium handling under physiological and pathological conditions as well as the newly identified contribution of the stress kinase JNK2 in CaMKIIδ-dependent SR Ca2+ abnormal mishandling. The new findings identifying dual roles of JNK2 in CaMKIIδ expression and activation are also discussed in this review.
Collapse
|
36
|
Ferrero G, Carpi S, Polini B, Pardini B, Nieri P, Impeduglia A, Grioni S, Tarallo S, Naccarati A. Intake of Natural Compounds and Circulating microRNA Expression Levels: Their Relationship Investigated in Healthy Subjects With Different Dietary Habits. Front Pharmacol 2021; 11:619200. [PMID: 33519486 PMCID: PMC7840481 DOI: 10.3389/fphar.2020.619200] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Diet has a strong influence on many physiological processes, which in turn have important implications on a variety of pathological conditions. In this respect, microRNAs (miRNAs), a class of small non-coding RNAs playing a relevant epigenetic role in controlling gene expression, may represent mediators between the dietary intake and the healthy status. Despite great advances in the field of nutri-epigenomics, it remains unclear how miRNA expression is modulated by the diet and, specifically, the intake of specific nutrients. We investigated the whole circulating miRNome by small RNA-sequencing performed on plasma samples of 120 healthy volunteers with different dietary habits (vegans, vegetarians, and omnivores). Dietary intakes of specific nutrients were estimated for each subject from the information reported in the food-frequency questionnaire previously validated in the EPIC study. We focused hereby on the intake of 23 natural compounds (NCs) of the classes of lipids, micro-elements, and vitamins. We identified 78 significant correlations (rho > 0.300, p-value < 0.05) among the estimated daily intake of 13 NCs and the expression levels of 58 plasma miRNAs. Overall, vitamin D, sodium, and vitamin E correlated with the largest number of miRNAs. All the identified correlations were consistent among the three dietary groups and 22 of them were confirmed as significant (p-value < 0.05) by age-, gender-, and body-mass index-adjusted Generalized Linear regression Model analysis. miR-23a-3p expression levels were related with different NCs including a significant positive correlation with sodium (rho = 0.377) and significant negative correlations with lipid-related NCs and vitamin E. Conversely, the estimated intake of vitamin D was negatively correlated with the expression of the highest number of circulating miRNAs, particularly miR-1277-5p (rho = −0.393) and miR-144-3p (rho = −0.393). Functional analysis of the targets of sodium intake-correlated miRNAs highlighted terms related to cardiac development. A similar approach on targets of those miRNAs correlated with vitamin D intake showed an enrichment in genes involved in hormone metabolisms, while the response to chronic inflammation was among the top enriched processes involving targets of miRNAs negatively related with vitamin E intake. Our findings show that nutrients through the habitual diet influence circulating miRNA profiles and highlight that this aspect must be considered in the nutri-epigenomic research.
Collapse
Affiliation(s)
- Giulio Ferrero
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy.,Department of Computer Science, University of Turin, Torino, Italy
| | - Sara Carpi
- Department of Pharmacy, University of Pisa, Pisa, Italy.,NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Pisa, Italy
| | | | - Barbara Pardini
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Torino, Italy
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Sara Grioni
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Sonia Tarallo
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Torino, Italy
| | - Alessio Naccarati
- Italian Institute for Genomic Medicine (IIGM), c/o IRCCS Candiolo, Torino, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Torino, Italy
| |
Collapse
|
37
|
Adiga D, Radhakrishnan R, Chakrabarty S, Kumar P, Kabekkodu SP. The Role of Calcium Signaling in Regulation of Epithelial-Mesenchymal Transition. Cells Tissues Organs 2020; 211:134-156. [PMID: 33316804 DOI: 10.1159/000512277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/13/2020] [Indexed: 11/19/2022] Open
Abstract
Despite substantial advances in the field of cancer therapeutics, metastasis is a significant challenge for a favorable clinical outcome. Epithelial to mesenchymal transition (EMT) is a process of acquiring increased motility, invasiveness, and therapeutic resistance by cancer cells for their sustained growth and survival. A plethora of intrinsic mechanisms and extrinsic microenvironmental factors drive the process of cancer metastasis. Calcium (Ca2+) signaling plays a critical role in dictating the adaptive metastatic cell behavior comprising of cell migration, invasion, angiogenesis, and intravasation. By modulating EMT, Ca2+ signaling can regulate the complexity and dynamics of events leading to metastasis. This review summarizes the role of Ca2+ signal remodeling in the regulation of EMT and metastasis in cancer.
Collapse
Affiliation(s)
- Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, India
| | - Prashant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India,
- Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, India,
| |
Collapse
|
38
|
Malison A, Arpanutud P, Keeratipibul S. Chicken foot broth byproduct: A new source for highly effective peptide-calcium chelate. Food Chem 2020; 345:128713. [PMID: 33310253 DOI: 10.1016/j.foodchem.2020.128713] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023]
Abstract
As a means of adding value, chicken foot broth byproduct can be processed to obtain calcium and bioactive peptides from the separated bones and meat residues. In this study, cleaned, dried, and powdered bones yielded 31.4 ± 0.6% calcium content. The meat residues were hydrolyzed to obtain over a hundred distinctive peptides, which were analyzed using LC-MS/MS and the SpirPep web-based tool. The peptides were rich in Glu, Asp, Lys, Gly and Leu, and also exhibited diverse bioactivities, among them primarily inhibition of dipeptidyl peptidase IV and angiotensin-converting enzyme. Calcium chelation assay determined the peptides to bind calcium at 235.7 ± 20.0 mg/g peptide-calcium chelate. Caco-2 cells treated with the chelate at calcium concentrations of 0-10 mM exhibited enhanced absorption relative to CaCl2. This demonstrates that calcium and chelating peptides generated from the same byproduct can produce peptide-calcium chelate, a potential ingredient in functional foods.
Collapse
Affiliation(s)
- Arichaya Malison
- Technopreneurship and Innovation Management Program, Graduate School Chulalongkorn University, Chamchuri 10 Building M1 Floor, Soi Chula 12, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Pornlert Arpanutud
- School of Liberal Arts, King Mongkut's University of Technology Thonburi, 126 Pracha-Uthit Road, Bangmod Thungkru, Bangkok 10140, Thailand
| | - Suwimon Keeratipibul
- Faculty of Science, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok 10330, Thailand.
| |
Collapse
|
39
|
Weisbrod D. Small and Intermediate Calcium Activated Potassium Channels in the Heart: Role and Strategies in the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:590534. [PMID: 33329039 PMCID: PMC7719780 DOI: 10.3389/fphys.2020.590534] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium-activated potassium channels are a heterogeneous family of channels that, despite their different biophysical characteristics, structures, and pharmacological signatures, play a role of transducer between the ubiquitous intracellular calcium signaling and the electric variations of the membrane. Although this family of channels was extensively described in various excitable and non-excitable tissues, an increasing amount of evidences shows their functional role in the heart. This review aims to focus on the physiological role and the contribution of the small and intermediate calcium-activated potassium channels in cardiac pathologies.
Collapse
|
40
|
Dridi H, Kushnir A, Zalk R, Yuan Q, Melville Z, Marks AR. Intracellular calcium leak in heart failure and atrial fibrillation: a unifying mechanism and therapeutic target. Nat Rev Cardiol 2020; 17:732-747. [PMID: 32555383 PMCID: PMC8362847 DOI: 10.1038/s41569-020-0394-8] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2020] [Indexed: 12/14/2022]
Abstract
Ca2+ is a fundamental second messenger in all cell types and is required for numerous essential cellular functions, including cardiac and skeletal muscle contraction. The intracellular concentration of free Ca2+ ([Ca2+]) is regulated primarily by ion channels, pumps (ATPases), exchangers and Ca2+-binding proteins. Defective regulation of [Ca2+] is found in a diverse spectrum of pathological states that affect all the major organs. In the heart, abnormalities in the regulation of cytosolic and mitochondrial [Ca2+] occur in heart failure (HF) and atrial fibrillation (AF), two common forms of heart disease and leading contributors to morbidity and mortality. In this Review, we focus on the mechanisms that regulate ryanodine receptor 2 (RYR2), the major sarcoplasmic reticulum (SR) Ca2+-release channel in the heart, how RYR2 becomes dysfunctional in HF and AF, and its potential as a therapeutic target. Inherited RYR2 mutations and/or stress-induced phosphorylation and oxidation of the protein destabilize the closed state of the channel, resulting in a pathological diastolic Ca2+ leak from the SR that both triggers arrhythmias and impairs contractility. On the basis of our increased understanding of SR Ca2+ leak as a shared Ca2+-dependent pathological mechanism in HF and AF, a new class of drugs developed in our laboratory, known as rycals, which stabilize RYR2 channels and prevent Ca2+ leak from the SR, are undergoing investigation in clinical trials.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Alexander Kushnir
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ran Zalk
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Zephan Melville
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
41
|
Fontaine D, Figiel S, Félix R, Kouba S, Fromont G, Mahéo K, Potier-Cartereau M, Chantôme A, Vandier C. Roles of endogenous ether lipids and associated PUFAs in the regulation of ion channels and their relevance for disease. J Lipid Res 2020; 61:840-858. [PMID: 32265321 PMCID: PMC7269763 DOI: 10.1194/jlr.ra120000634] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Indexed: 12/16/2022] Open
Abstract
Ether lipids (ELs) are lipids characterized by the presence of either an ether linkage (alkyl lipids) or a vinyl ether linkage [i.e., plasmalogens (Pls)] at the sn1 position of the glycerol backbone, and they are enriched in PUFAs at the sn2 position. In this review, we highlight that ELs have various biological functions, act as a reservoir for second messengers (such as PUFAs) and have roles in many diseases. Some of the biological effects of ELs may be associated with their ability to regulate ion channels that control excitation-contraction/secretion/mobility coupling and therefore cell physiology. These channels are embedded in lipid membranes, and lipids can regulate their activities directly or indirectly as second messengers or by incorporating into membranes. Interestingly, ELs and EL-derived PUFAs have been reported to play a key role in several pathologies, including neurological disorders, cardiovascular diseases, and cancers. Investigations leading to a better understanding of their mechanisms of action in pathologies have opened a new field in cancer research. In summary, newly identified lipid regulators of ion channels, such as ELs and PUFAs, may represent valuable targets to improve disease diagnosis and advance the development of new therapeutic strategies for managing a range of diseases and conditions.
Collapse
Affiliation(s)
- Delphine Fontaine
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sandy Figiel
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Romain Félix
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sana Kouba
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Gaëlle Fromont
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Department of Pathology, CHRU Bretonneau, F-37044 Tours CEDEX 9, France
| | - Karine Mahéo
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | | | - Aurélie Chantôme
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | - Christophe Vandier
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France. mailto:
| |
Collapse
|
42
|
Essandoh K, Philippe JM, Jenkins PM, Brody MJ. Palmitoylation: A Fatty Regulator of Myocardial Electrophysiology. Front Physiol 2020; 11:108. [PMID: 32140110 PMCID: PMC7042378 DOI: 10.3389/fphys.2020.00108] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/30/2020] [Indexed: 01/02/2023] Open
Abstract
Regulation of cardiac physiology is well known to occur through the action of kinases that reversibly phosphorylate ion channels, calcium handling machinery, and signaling effectors. However, it is becoming increasingly apparent that palmitoylation or S-acylation, the post-translational modification of cysteines with saturated fatty acids, plays instrumental roles in regulating the localization, activity, stability, sorting, and function of numerous proteins, including proteins known to have essential functions in cardiomyocytes. However, the impact of this modification on cardiac physiology requires further investigation. S-acylation is catalyzed by the zDHHC family of S-acyl transferases that localize to intracellular organelle membranes or the sarcolemma. Recent work has begun to uncover functions of S-acylation in the heart, particularly in the regulation of cardiac electrophysiology, including modification of the sodium-calcium exchanger, phospholemman and the cardiac sodium pump, as well as the voltage-gated sodium channel. Elucidating the regulatory functions of zDHHC enzymes in cardiomyocytes and determination of how S-acylation is altered in the diseased heart will shed light on how these modifications participate in cardiac pathogenesis and potentially identify novel targets for the treatment of cardiovascular disease. Indeed, proteins with critical signaling roles in the heart are also S-acylated, including receptors and G-proteins, yet the dynamics and functions of these modifications in myocardial physiology have not been interrogated. Here, we will review what is known about zDHHC enzymes and substrate S-acylation in myocardial physiology and highlight future areas of investigation that will uncover novel functions of S-acylation in cardiac homeostasis and pathophysiology.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Julie M Philippe
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Paul M Jenkins
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States.,Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
43
|
Sillanpää JK, Cardoso JCDR, Félix RC, Anjos L, Power DM, Sundell K. Dilution of Seawater Affects the Ca 2 + Transport in the Outer Mantle Epithelium of Crassostrea gigas. Front Physiol 2020; 11:1. [PMID: 32038307 PMCID: PMC6987452 DOI: 10.3389/fphys.2020.00001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/01/2020] [Indexed: 12/18/2022] Open
Abstract
Varying salinities of coastal waters are likely to affect the physiology and ion transport capabilities of calcifying marine organisms such as bivalves. To investigate the physiological effect of decreased environmental salinity in bivalves, adult oysters (Crassostrea gigas) were exposed for 14 days to 50% seawater (14) and the effects on mantle ion transport, electrophysiology and the expression of Ca2+ transporters and channels relative to animals maintained in full strength sea water (28) was evaluated. Exposure of oysters to a salinity of 14 decreased the active mantle transepithelial ion transport and specifically affected Ca2+ transfer. Gene expression of the Na+/K+-ATPase and the sarco(endo)plasmic reticulum Ca2+-ATPase was decreased whereas the expression of the T-type voltage-gated Ca channel and the Na+/Ca2+-exchanger increased compared to animals maintained in full SW. The results indicate that decreased environmental salinities will most likely affect not only osmoregulation but also bivalve biomineralization and shell formation.
Collapse
Affiliation(s)
- J Kirsikka Sillanpää
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Joao Carlos Dos Reis Cardoso
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro, Portugal
| | - Rute Castelo Félix
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro, Portugal
| | - Liliana Anjos
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro, Portugal
| | - Deborah Mary Power
- Comparative Endocrinology and Integrative Biology, Centre of Marine Sciences, Universidade do Algarve, Faro, Portugal
| | - Kristina Sundell
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Gök C, Fuller W. Regulation of NCX1 by palmitoylation. Cell Calcium 2020; 86:102158. [PMID: 31935590 DOI: 10.1016/j.ceca.2019.102158] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/28/2019] [Accepted: 12/29/2019] [Indexed: 11/17/2022]
Abstract
Palmitoylation (S-acylation) is the reversible conjugation of a fatty acid (usually C16 palmitate) to intracellular cysteine residues of proteins via a thioester linkage. Palmitoylation anchors intracellular regions of proteins to membranes because the palmitoylated cysteine is recruited to the lipid bilayer. NCX1 is palmitoylated at a single cysteine in its large regulatory intracellular loop. The presence of an amphipathic α-helix immediately adjacent to the NCX1 palmitoylation site is required for NCX1 palmitoylation. The NCX1 palmitoylation site is conserved through most metazoan phlya. Although palmitoylation does not regulate the normal forward or reverse ion transport modes of NCX1, NCX1 palmitoylation is required for its inactivation: sodium-dependent inactivation and inactivation by PIP2 depletion are significantly impaired for unpalmitoylatable NCX1. Here we review the role of palmitoylation in regulating NCX1 activity, and highlight future questions that must be addressed to fully understand the importance of this regulatory mechanism for sodium and calcium transport in cardiac muscle.
Collapse
Affiliation(s)
- Caglar Gök
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - William Fuller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK. https://twitter.com@FullerLabGlas
| |
Collapse
|
45
|
Al Kury LT. Calcium Homeostasis in Ventricular Myocytes of Diabetic Cardiomyopathy. J Diabetes Res 2020; 2020:1942086. [PMID: 33274235 PMCID: PMC7683117 DOI: 10.1155/2020/1942086] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder commonly characterized by high blood glucose levels, resulting from defects in insulin production or insulin resistance, or both. DM is a leading cause of mortality and morbidity worldwide, with diabetic cardiomyopathy as one of its main complications. It is well established that cardiovascular complications are common in both types of diabetes. Electrical and mechanical problems, resulting in cardiac contractile dysfunction, are considered as the major complications present in diabetic hearts. Inevitably, disturbances in the mechanism(s) of Ca2+ signaling in diabetes have implications for cardiac myocyte contraction. Over the last decade, significant progress has been made in outlining the mechanisms responsible for the diminished cardiac contractile function in diabetes using different animal models of type I diabetes mellitus (TIDM) and type II diabetes mellitus (TIIDM). The aim of this review is to evaluate our current understanding of the disturbances of Ca2+ transport and the role of main cardiac proteins involved in Ca2+ homeostasis in the diabetic rat ventricular cardiomyocytes. Exploring the molecular mechanism(s) of altered Ca2+ signaling in diabetes will provide an insight for the identification of novel therapeutic approaches to improve the heart function in diabetic patients.
Collapse
Affiliation(s)
- Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, UAE
| |
Collapse
|
46
|
Mussel M, Horkay F. Experimental Evidence for Universal Behavior of Ion-Induced Volume Phase Transition in Sodium Polyacrylate Gels. J Phys Chem Lett 2019; 10:7831-7835. [PMID: 31804832 PMCID: PMC8243402 DOI: 10.1021/acs.jpclett.9b03126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Introduction of high valence counterions into polyelectrolyte gels results in a reversible volume phase transition. In the present work new experimental results are reported for the volume transition induced by calcium/sodium exchange in sodium polyacrylate gels. The effects of cross-link density, concentration of ionized groups on the network chains, composition of the equilibrium salt solution containing both mono- and divalent cations, and temperature on the swelling degree of these gels are systematically investigated. It is demonstrated that the normalized swelling data fall on a master curve, indicating that the ion-exchange-induced volume transition exhibits universal behavior in sodium polyacrylate gels. Model calculations made on the basis of the classical Flory-Rehner theory are in reasonable agreement with the measured dependencies.
Collapse
|
47
|
Zhao J, Xu T, Zhou Y, Zhou Y, Xia Y, Li D. B-type natriuretic peptide and its role in altering Ca 2+-regulatory proteins in heart failure-mechanistic insights. Heart Fail Rev 2019; 25:861-871. [PMID: 31820203 DOI: 10.1007/s10741-019-09883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heart failure (HF) is a worldwide disease with high levels of morbidity and mortality. The pathogenesis of HF is complicated and involves imbalances in hormone and electrolyte. B-type natriuretic peptide (BNP) has served as a biomarker of HF severity, and in recent years, it has been used to treat the disease, thanks to its cardio-protective effects, such as diuresis, natriuresis, and vasodilatation. In stage C/D HF, symptoms are severe despite elevated BNP. Disturbances in Ca2+ homeostasis are often a dominating feature of the disease, causing Ca2+-regulatory protein dysfunction, including reduced expression and activity of sarcoplasmic reticulum Ca2+-ATPase2a (SERCA2a), impaired ryanodine receptors (RYRs) function, intensive Na+-Ca2+ exchanger (NCX), and downregulation of S100A1. The relationship between natriuretic peptides (NPs) and Ca2+-regulatory proteins has been widely studied and represents important mechanisms in the etiology of HF. In this review, we present evidence that BNP may regulate Ca2+-regulatory proteins, in particular, suppressing SERCA2a and S100A1 expression. However, relationships between BNP and other Ca2+-regulatory proteins remain vague.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yao Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - You Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yong Xia
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
48
|
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: 11] [Impact Index Per Article: 1.8] [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
|
49
|
Sherman WF, Grosberg A. Exploring cardiac form and function: A length-scale computational biology approach. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2019; 12:e1470. [PMID: 31793215 DOI: 10.1002/wsbm.1470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/08/2019] [Accepted: 11/06/2019] [Indexed: 01/14/2023]
Abstract
The ability to adequately pump blood throughout the body is the result of tightly regulated feedback mechanisms that exist across many spatial scales in the heart. Diseases which impede the function at any one of the spatial scales can cause detrimental cardiac remodeling and eventual heart failure. An overarching goal of cardiac research is to use engineered heart tissue in vitro to study the physiology of diseased heart tissue, develop cell replacement therapies, and explore drug testing applications. A commonality within the field is to manipulate the flow of mechanical signals across the various spatial scales to direct self-organization and build functional tissue. Doing so requires an understanding of how chemical, electrical, and mechanical cues can be used to alter the cellular microenvironment. We discuss how mathematical models have been used in conjunction with experimental techniques to explore various structure-function relations that exist across numerous spatial scales. We highlight how a systems biology approach can be employed to recapitulate in vivo characteristics in vitro at the tissue, cell, and subcellular scales. Specific focus is placed on the interplay between experimental and theoretical approaches. Various modeling methods are showcased to demonstrate the breadth and power afforded to the systems biology approach. An overview of modeling methodologies exemplifies how the strengths of different scientific disciplines can be used to supplement and/or inspire new avenues of experimental exploration. This article is categorized under: Models of Systems Properties and Processes > Mechanistic Models Models of Systems Properties and Processes > Cellular Models Models of Systems Properties and Processes > Organ, Tissue, and Physiological Models.
Collapse
Affiliation(s)
- William F Sherman
- Center for Complex Biological Systems, University of California Irvine, Irvine, California.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California
| | - Anna Grosberg
- Center for Complex Biological Systems, University of California Irvine, Irvine, California.,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, California.,Department of Biomedical Engineering, University of California Irvine, Irvine, California.,Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California.,NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, California
| |
Collapse
|
50
|
Primessnig U, Bracic T, Levijoki J, Otsomaa L, Pollesello P, Falcke M, Pieske B, Heinzel FR. Long-term effects of Na + /Ca 2+ exchanger inhibition with ORM-11035 improves cardiac function and remodelling without lowering blood pressure in a model of heart failure with preserved ejection fraction. Eur J Heart Fail 2019; 21:1543-1552. [PMID: 31762174 DOI: 10.1002/ejhf.1619] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 08/26/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is increasingly common but there is currently no established pharmacological therapy. We hypothesized that ORM-11035, a novel specific Na+ /Ca2+ exchanger (NCX) inhibitor, improves cardiac function and remodelling independent of effects on arterial blood pressure in a model of cardiorenal HFpEF. METHODS AND RESULTS Rats were subjected to subtotal nephrectomy (NXT) or sham operation. Eight weeks after intervention, treatment for 16 weeks with ORM-11035 (1 mg/kg body weight) or vehicle was initiated. At 24 weeks, blood pressure measurements, echocardiography and pressure-volume loops were performed. Contractile function, Ca2+ transients and NCX-mediated Ca2+ extrusion were measured in isolated ventricular cardiomyocytes. NXT rats (untreated) showed a HFpEF phenotype with left ventricular (LV) hypertrophy, LV end-diastolic pressure (LVEDP) elevation, increased brain natriuretic peptide (BNP) levels, preserved ejection fraction and pulmonary congestion. In cardiomyocytes from untreated NXT rats, early relaxation was prolonged and NCX-mediated Ca2+ extrusion was decreased. Chronic treatment with ORM-11035 significantly reduced LV hypertrophy and cardiac remodelling without lowering systolic blood pressure. LVEDP [14 ± 3 vs. 9 ± 2 mmHg; NXT (n = 12) vs. NXT + ORM (n = 12); P = 0.0002] and BNP levels [71 ± 12 vs. 49 ± 11 pg/mL; NXT (n = 12) vs. NXT + ORM (n = 12); P < 0.0001] were reduced after ORM treatment. LV cardiomyocytes from ORM-treated rats showed improved active relaxation and diastolic cytosolic Ca2+ decay as well as restored NCX-mediated Ca2+ removal, indicating NCX modulation with ORM-11035 as a promising target in the treatment of HFpEF. CONCLUSION Chronic inhibition of NCX with ORM-11035 significantly attenuated cardiac remodelling and diastolic dysfunction without lowering systemic blood pressure in this model of HFpEF. Therefore, long-term treatment with selective NCX inhibitors such as ORM-11035 should be evaluated further in the treatment of heart failure.
Collapse
Affiliation(s)
- Uwe Primessnig
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Department of Cardiology, Medical University of Graz, Graz, Austria.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Taja Bracic
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | | | | | | | - Martin Falcke
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Physics, Humboldt Universität, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| |
Collapse
|