1
|
Alquisiras-Burgos I, Peralta-Arrieta I, Espinoza-Rojo M, Salazar-Salgado A, Antonino-Olguín I, Sánchez-Mendoza A, Sánchez-Aguilar M, Ruiz-Tachiquín ME, Valdez-Salazar HA, Ortiz-Plata A, Franco-Pérez J, Hernández-Cruz A, Aguilera P. Expression of SUR1 isoforms in the brain and heart after ischemia/reperfusion. Front Mol Neurosci 2025; 18:1536409. [PMID: 40313402 PMCID: PMC12043708 DOI: 10.3389/fnmol.2025.1536409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
The sulfonylurea receptor 1 (SUR1) has been classified as a member of the adenosine triphosphate (ATP)-binding cassette (ABC) transporter superfamily. SUR1, unlike the classic ABC transporters, assembles with Kir6.2, forming KATP channels to regulate the flux of potassium ions. In the central nervous system, SUR1 is weakly expressed in some brain regions but is induced by pathological conditions in the different cell types of the neurovascular unit. Therefore, we first analyzed the expression of SUR1 in various rat tissues and brain regions to identify SUR1 isoforms and their mRNA exon composition under physiological conditions. Later, we focused on the SUR1 expression in the brain and heart after ischemia/reperfusion. We observed two SUR1 isoforms (170 and 60-75 kDa) abundantly expressed in most rat tissues, except for the testis and brain, where basal expression of these isoforms was relatively low and exhibit a band of 100 kDa. Every exons coding for the functional domains of SUR1 mRNA were amplified from the tissues and brain regions analyzed. Results from in vitro and in vivo experiments indicated that SUR1 isoforms previously identified (170 and 60-75 kDa) were dramatically overexpressed in the brain after middle cerebral artery occlusion followed by reperfusion. In contrast, myocardial infarction followed by reperfusion significantly reduced SUR1 isoform expression in the heart. This study demonstrates the expression of at least two SUR1 isoforms in various tissues and suggests that ischemic processes may differentially regulate SUR1 expression depending on the tissue injured.
Collapse
Affiliation(s)
- Iván Alquisiras-Burgos
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Irlanda Peralta-Arrieta
- Laboratorio de Transducción de Señales, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Mónica Espinoza-Rojo
- Laboratorio de Biología Molecular y Genómica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo, Mexico
| | - Alejandro Salazar-Salgado
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Iván Antonino-Olguín
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
- Laboratorio de Biología Molecular y Genómica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo, Mexico
| | - Alicia Sánchez-Mendoza
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| | - María Sánchez-Aguilar
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| | - Martha-Eugenia Ruiz-Tachiquín
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Hilda-Alicia Valdez-Salazar
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Alma Ortiz-Plata
- Laboratorio de Patología Experimental, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Javier Franco-Pérez
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Arturo Hernández-Cruz
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Penélope Aguilera
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| |
Collapse
|
2
|
Choe JC, Oh JH, Lee HC, Lee JW, Park TS, Park JH, Kim LE, Kim MS, Ahn J, Park JS, Lee HW, Choi JH, Cha KS. The effect of nicorandil on cardiac function and clinical outcomes in ST-segment elevation myocardial infarction patients undergoing primary percutaneous coronary intervention: a randomised trial. Acta Cardiol 2023; 78:880-888. [PMID: 36942830 DOI: 10.1080/00015385.2022.2129592] [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: 04/06/2022] [Accepted: 09/05/2022] [Indexed: 03/23/2023]
Abstract
BACKGROUND We investigated the effect of nicorandil on infarct size, cardiac function assessed by cardiac magnetic resonance imaging (CMR) and outcomes in ST-segment elevation myocardial infarction (STEMI) patients undergoing primary percutaneous coronary intervention (PCI). METHODS In a prospective, randomised, controlled trial, 83 patients with STEMI receiving primary PCI were randomised into the nicorandil (n = 40) or placebo (n = 43) groups. Nicorandil was administered in the emergency room before primary PCI as an intravenous bolus of 4 mg followed by a continuous infusion of 6 mg/h for 24 h and as 2-mg intracoronary injections prior to balloon dilatation and coronary stenting. Nicorandil was continued orally at 10-20 mg/d for 6 months. Infarct size and cardiac function were measured by CMR at 5 d and 6 months after primary PCI. Furthermore, major adverse cardiac events (MACEs) including all-cause death, nonfatal myocardial infarction (MI), any revascularisation, stroke, and definite/probable stent thrombosis (ST) were compared. RESULTS There were no significant differences in baseline clinical characteristics between the groups. Infarct size at baseline and 6 months as well as infarct size changes during 6 months as measured by CMR were similar between the groups. Similarly, other CMR parameters were comparable at baseline and 6 months between the groups. MACEs occurred in four patients (4.8%) during 6 months. No significant difference in the risk of MACEs was observed between the groups. CONCLUSIONS Treatment with nicorandil for 6 months after primary PCI was not associated with any improvement in infarct size, CMR-determined cardiac function, and outcomes in STEMI patients.
Collapse
Affiliation(s)
- Jeong Cheon Choe
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jun-Hyok Oh
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Han Cheol Lee
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Ji Won Lee
- Department of Radiology, Pusan National University Hospital, Busan, Republic of Korea
| | - Tae Sik Park
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jong Ha Park
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - La Eun Kim
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Min Su Kim
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Hospital, Busan, Republic of Korea
| | - Jinhee Ahn
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jin Sup Park
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hye Won Lee
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Jung Hyun Choi
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Kwang Soo Cha
- Department of Internal Medicine, Division of Cardiology, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
3
|
Caruba T, Charles-Nelson A, Alexander JH, Mehta RH, Heringlake M, Toller W, Yavchitz A, Sabatier B, Chatellier G, Cholley B. Prophylactic levosimendan in patients with low ejection fraction undergoing coronary artery bypass grafting: A pooled analysis of two multicentre randomised controlled trials. Anaesth Crit Care Pain Med 2022; 41:101107. [PMID: 35643391 DOI: 10.1016/j.accpm.2022.101107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To assess the effect of preoperative levosimendan on mortality at Day-90 in patients with left ventricular ejection fraction (LVEF) ≤ 40%, and to investigate a possible differential effect between patients undergoing isolated coronary artery bypass grafting (CABG) versus CABG combined with valve replacement surgery. DESIGN Pooled analysis of two multicentre randomised controlled trials (RCT) investigating prophylactic levosimendan versus placebo prior to CABG surgery on mortality at Day-90 in patients with LVEF ≤ 40%. A meta-analysis of all RCT investigating the same issue was also conducted. RESULTS A cohort of 1084 patients (809 isolated CABG, and 275 combined surgery) resulted from the merging of LEVO-CTS and LICORN databases. Seventy-two patients were dead at day 90. The mortality at day 90 was not different between levosimendan and placebo (Hazard Ratio (HR): 0.73, 95% CI: 0.41-1.28, p = 0.27). However, there was a significant interaction between the type of surgery and the study drug (p = 0.004). We observed a decrease in mortality at day 90 in the isolated CABG subgroup (HR: 0.39, 95% CI: 0.19-0.82, p = 0.013), but not in the combined surgery subgroup (HR: 1.73, 95% CI: 0.77-3.92, p = 0.19). The meta-analysis of 6 RCT involving 1441 patients confirmed the differential effect on mortality at day 30 between the 2 subgroups. CONCLUSIONS Preoperative levosimendan did not reduce mortality in a mixed surgical population with LV dysfunction. However, the subgroup of patients undergoing isolated CABG had a reduction in mortality at day 90, whereas there was no significant effect in combined surgery patients. This finding requires confirmation with a specific prospective trial.
Collapse
Affiliation(s)
- Thibaut Caruba
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | | | - John H Alexander
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Rajendra H Mehta
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthias Heringlake
- Dept. of Anaesthesiology and Intensive Care Medicine, Heart- and Diabetes Center, Karlsburg, Germany
| | | | - Amelie Yavchitz
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Brigitte Sabatier
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France; Inserm UMR_1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris Cité, F-75006 Paris, France and Inria, HeKA, PariSantéCampus, Paris, France
| | - Gilles Chatellier
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France; Université de Paris, F-75006 Paris, France
| | - Bernard Cholley
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France; Université de Paris, INSERM UMR_S 1140 "Innovations Thérapeutiques en Hémostase", F-75006 Paris, France.
| |
Collapse
|
4
|
Naryzhnaya NV, Maslov LN, Derkachev IA, Fu F. The Significance of NO-Synthase, Reactive Oxygen Species, Kinases and KATP-Channels in the Development of the Infarct-Limiting Effect of Adaptation to Hypoxia. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Naryzhnaya NV, Maslov LN, Derkachev IA, Ma H, Zhang Y, Prasad NR, Singh N, Fu F, Pei JM, Sarybaev A, Sydykov A. The effect of adaptation to hypoxia on cardiac tolerance to ischemia/reperfusion. J Biomed Res 2022:1-25. [PMID: 37183617 PMCID: PMC10387748 DOI: 10.7555/jbr.36.20220125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The acute myocardial infarction (AMI) and sudden cardiac death (SCD), both associated with acute cardiac ischemia, are one of the leading causes of adult death in economically developed countries. The development of new approaches for the treatment and prevention of AMI and SCD remains the highest priority for medicine. A study on the cardiovascular effects of chronic hypoxia (CH) may contribute to the development of these methods. Chronic hypoxia exerts both positive and adverse effects. The positive effects are the infarct-reducing, vasoprotective, and antiarrhythmic effects, which can lead to the improvement of cardiac contractility in reperfusion. The adverse effects are pulmonary hypertension and right ventricular hypertrophy. This review presents a comprehensive overview of how CH enhances cardiac tolerance to ischemia/reperfusion. It is an in-depth analysis of the published data on the underlying mechanisms, which can lead to future development of the cardioprotective effect of CH. A better understanding of the CH-activated protective signaling pathways may contribute to new therapeutic approaches in an increase of cardiac tolerance to ischemia/reperfusion.
Collapse
|
6
|
Abstract
K+ channels enable potassium to flow across the membrane with great selectivity. There are four K+ channel families: voltage-gated K (Kv), calcium-activated (KCa), inwardly rectifying K (Kir), and two-pore domain potassium (K2P) channels. All four K+ channels are formed by subunits assembling into a classic tetrameric (4x1P = 4P for the Kv, KCa, and Kir channels) or tetramer-like (2x2P = 4P for the K2P channels) architecture. These subunits can either be the same (homomers) or different (heteromers), conferring great diversity to these channels. They share a highly conserved selectivity filter within the pore but show different gating mechanisms adapted for their function. K+ channels play essential roles in controlling neuronal excitability by shaping action potentials, influencing the resting membrane potential, and responding to diverse physicochemical stimuli, such as a voltage change (Kv), intracellular calcium oscillations (KCa), cellular mediators (Kir), or temperature (K2P).
Collapse
|
7
|
Mitochondrial osmoregulation in evolution, cation transport and metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148368. [PMID: 33422486 DOI: 10.1016/j.bbabio.2021.148368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 11/24/2022]
Abstract
This review provides a retrospective on the role of osmotic regulation in the process of eukaryogenesis. Specifically, it focuses on the adjustments which must have been made by the original colonizing α-proteobacteria that led to the evolution of modern mitochondria. We focus on the cations that are fundamentally involved in volume determination and cellular metabolism and define the transporter landscape in relation to these ions in mitochondria as we know today. We provide analysis on how the cations interplay and together maintain osmotic balance that allows for effective ATP synthesis in the organelle.
Collapse
|
8
|
Hausenloy DJ, Schulz R, Girao H, Kwak BR, De Stefani D, Rizzuto R, Bernardi P, Di Lisa F. Mitochondrial ion channels as targets for cardioprotection. J Cell Mol Med 2020; 24:7102-7114. [PMID: 32490600 PMCID: PMC7339171 DOI: 10.1111/jcmm.15341] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/31/2020] [Accepted: 04/12/2020] [Indexed: 12/14/2022] Open
Abstract
Acute myocardial infarction (AMI) and the heart failure (HF) that often result remain the leading causes of death and disability worldwide. As such, new therapeutic targets need to be discovered to protect the myocardium against acute ischaemia/reperfusion (I/R) injury in order to reduce myocardial infarct (MI) size, preserve left ventricular function and prevent the onset of HF. Mitochondrial dysfunction during acute I/R injury is a critical determinant of cell death following AMI, and therefore, ion channels in the inner mitochondrial membrane, which are known to influence cell death and survival, provide potential therapeutic targets for cardioprotection. In this article, we review the role of mitochondrial ion channels, which are known to modulate susceptibility to acute myocardial I/R injury, and we explore their potential roles as therapeutic targets for reducing MI size and preventing HF following AMI.
Collapse
Affiliation(s)
- Derek J. Hausenloy
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart Research Institute SingaporeNational Heart CentreSingaporeSingapore
- Yong Loo Lin School of MedicineNational University SingaporeSingaporeSingapore
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular Research CenterCollege of Medical and Health SciencesAsia UniversityTaichung CityTaiwan
| | - Rainer Schulz
- Institute of PhysiologyJustus‐Liebig University GiessenGiessenGermany
| | - Henrique Girao
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of MedicineUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
- Clinical Academic Centre of CoimbraCACCCoimbraPortugal
| | - Brenda R. Kwak
- Department of Pathology and ImmunologyUniversity of GenevaGenevaSwitzerland
| | - Diego De Stefani
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Rosario Rizzuto
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Paolo Bernardi
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- CNR Neuroscience InstitutePadovaItaly
| | - Fabio Di Lisa
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- CNR Neuroscience InstitutePadovaItaly
| |
Collapse
|
9
|
Walsh KB. Screening Technologies for Inward Rectifier Potassium Channels: Discovery of New Blockers and Activators. SLAS DISCOVERY 2020; 25:420-433. [PMID: 32292089 DOI: 10.1177/2472555220905558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
K+ channels play a critical role in maintaining the normal electrical activity of excitable cells by setting the cell resting membrane potential and by determining the shape and duration of the action potential. In nonexcitable cells, K+ channels establish electrochemical gradients necessary for maintaining salt and volume homeostasis of body fluids. Inward rectifier K+ (Kir) channels typically conduct larger inward currents than outward currents, resulting in an inwardly rectifying current versus voltage relationship. This property of inward rectification results from the voltage-dependent block of the channels by intracellular polyvalent cations and makes these channels uniquely designed for maintaining the resting potential near the K+ equilibrium potential (EK). The Kir family of channels consist of seven subfamilies of channels (Kir1.x through Kir7.x) that include the classic inward rectifier (Kir2.x) channel, the G-protein-gated inward rectifier K+ (GIRK) (Kir3.x), and the adenosine triphosphate (ATP)-sensitive (KATP) (Kir 6.x) channels as well as the renal Kir1.1 (ROMK), Kir4.1, and Kir7.1 channels. These channels not only function to regulate electrical/electrolyte transport activity, but also serve as effector molecules for G-protein-coupled receptors (GPCRs) and as molecular sensors for cell metabolism. Of significance, Kir channels represent promising pharmacological targets for treating a number of clinical conditions, including cardiac arrhythmias, anxiety, chronic pain, and hypertension. This review provides a brief background on the structure, function, and pharmacology of Kir channels and then focuses on describing and evaluating current high-throughput screening (HTS) technologies, such as membrane potential-sensitive fluorescent dye assays, ion flux measurements, and automated patch clamp systems used for Kir channel drug discovery.
Collapse
Affiliation(s)
- Kenneth B Walsh
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
10
|
Madreiter‐Sokolowski CT, Ramadani‐Muja J, Ziomek G, Burgstaller S, Bischof H, Koshenov Z, Gottschalk B, Malli R, Graier WF. Tracking intra- and inter-organelle signaling of mitochondria. FEBS J 2019; 286:4378-4401. [PMID: 31661602 PMCID: PMC6899612 DOI: 10.1111/febs.15103] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/19/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
Abstract
Mitochondria are as highly specialized organelles and masters of the cellular energy metabolism in a constant and dynamic interplay with their cellular environment, providing adenosine triphosphate, buffering Ca2+ and fundamentally contributing to various signaling pathways. Hence, such broad field of action within eukaryotic cells requires a high level of structural and functional adaptation. Therefore, mitochondria are constantly moving and undergoing fusion and fission processes, changing their shape and their interaction with other organelles. Moreover, mitochondrial activity gets fine-tuned by intra- and interorganelle H+ , K+ , Na+ , and Ca2+ signaling. In this review, we provide an up-to-date overview on mitochondrial strategies to adapt and respond to, as well as affect, their cellular environment. We also present cutting-edge technologies used to track and investigate subcellular signaling, essential to the understanding of various physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Corina T. Madreiter‐Sokolowski
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
- Department of Health Sciences and TechnologyETH ZurichSchwerzenbachSwitzerland
| | - Jeta Ramadani‐Muja
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Gabriela Ziomek
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Sandra Burgstaller
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Helmut Bischof
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Zhanat Koshenov
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
- BioTechMedGrazAustria
| | - Wolfgang F. Graier
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
- BioTechMedGrazAustria
| |
Collapse
|
11
|
Blignaut M, Espach Y, van Vuuren M, Dhanabalan K, Huisamen B. Revisiting the Cardiotoxic Effect of Chloroquine. Cardiovasc Drugs Ther 2019; 33:1-11. [PMID: 30635818 DOI: 10.1007/s10557-018-06847-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Cardiotoxicity is a well-known side effect of chloroquine. Several studies have proposed chloroquine as a potential anti-diabetic treatment but do not address this problem. The current study investigated the effect of ex vivo chloroquine treatment on (1) heart function and glucose uptake, (2) mitochondrial function and (3) in vivo treatment on heart function. METHODS Control or obese male Wistar rats were used throughout. Dose responses of increasing chloroquine concentrations versus vehicle on cardiac function were measured using isolated, Langendorff-perfused hearts whilst glucose uptake and cell viability were determined in ventricular cardiomyocytes. Mitochondrial function was assessed with a Clark-type oxygraph (Hansatech) after ex vivo perfusion with 30 μM chloroquine versus vehicle. Animals were treated orally with 5 mg/kg/day chloroquine for 6 weeks. RESULTS Acute chloroquine treatment of 10 μM was sufficient to significantly decrease heart function (p < 0.05) whilst 30 μM significantly reduced heart rate (p < 0.05). Chloroquine became toxic to isolated cardiomyocytes at high concentrations (100 μM), and had no effect on cardiomyocyte glucose uptake. Ex vivo treatment did not affect mitochondrial function, but chronic low-dose in vivo chloroquine treatment significantly decreased aortic output and total work in hearts (p < 0.005). CONCLUSION Low and intermediate chloroquine doses administered either chronically or acutely are sufficient to result in myocardial dysfunction.
Collapse
Affiliation(s)
- Marguerite Blignaut
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Yolandi Espach
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Mignon van Vuuren
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Karthik Dhanabalan
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Barbara Huisamen
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa. .,South African Medical Research Council, Biomedical Research and Innovation Platform, Tygerberg, South Africa.
| |
Collapse
|
12
|
Inward rectifier potassium (Kir) channels mediate salivary gland function and blood feeding in the lone star tick, Amblyomma americanum. PLoS Negl Trop Dis 2019; 13:e0007153. [PMID: 30730880 PMCID: PMC6382211 DOI: 10.1371/journal.pntd.0007153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/20/2019] [Accepted: 01/13/2019] [Indexed: 02/06/2023] Open
Abstract
Background Tick feeding causes extreme morbidity and mortality to humans through transmission of pathogens and causes severe economic losses to the agricultural industry by reducing livestock yield. Salivary gland secretions are essential for tick feeding and thus, reducing or preventing saliva secretions into the vertebrate host is likely to reduce feeding and hinder pathogen life cycles. Unfortunately, the membrane physiology of tick salivary glands is underexplored and this gap in knowledge limits the development of novel therapeutics for inducing cessation of tick feeding. Methodology We studied the influence of inward rectifier potassium (Kir) channel subtypes to the functional capacity of the isolated tick salivary gland through the use of a modified Ramsay assay. The secreted saliva was subsequently used for quantification of the elemental composition of the secreted saliva after the glands were exposed to K+ channel modulators as a measure of osmoregulatory capacity. Lastly, changes to blood feeding behavior and mortality were measured with the use of a membrane feeding system. Principal findings In this study, we characterized the fundamental role of Kir channel subtypes in tick salivary gland function and provide evidence that pharmacological inhibition of these ion channels reduces the secretory activity of the Amblyomma americanum salivary gland. The reduced secretory capacity of the salivary gland was directly correlated with a dramatic reduction of blood ingestion during feeding. Further, exposure to small-molecule modulators of Kir channel subtypes induced mortality to ticks that is likely resultant from an altered osmoregulatory capacity. Conclusions Our data contribute to understanding of tick salivary gland function and could guide future campaigns aiming to develop chemical or reverse vaccinology technologies to reduce the worldwide burden of tick feeding and tick-vectored pathogens. Tick feeding results in negative health and economic consequences worldwide and there has been continued interest in the development of products with novel mechanisms of action for control of tick populations. Kir channels have been shown to be a significant ion conductance pathway in arthropods and are critical for proper functioning of multiple biological processes. Previous work on insect Kir channels has focused on their physiological roles in renal system of mosquitoes and the data suggest that these channels represent a viable pathway to induce renal failure that leads to mortality. Based on the functional and cellular similarities of arthropod salivary glands and Malpighian tubules, we hypothesized that Kir channels constitute a critical conductance pathway within arthropod salivary glands and inhibition of this pathway will preclude feeding. Data presented in this study show that pharmacological modulators of Kir channels elicited a significant reduction in the fluid and ion secretory activity of tick salivary glands that resulted in reduced feeding, altered osmoregulation, and lead to mortality. These data could guide the future development of novel acaricides, RNAi, or genetically modified ticks to mitigate health and economic damages resulting from their feeding. Further, these data indicate a conserved function of Kir channels within multiple tissues of taxonomically diverse organisms, such as ticks and humans.
Collapse
|
13
|
Joshi S, Jarajapu YPR. Mitochondrial depolarization stimulates vascular repair-relevant functions of CD34 + cells via reactive oxygen species-induced nitric oxide generation. Br J Pharmacol 2018; 176:4373-4387. [PMID: 30367728 DOI: 10.1111/bph.14529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/23/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE CD34+ haematopoietic stem/progenitor cells have revascularization potential and are now being tested for the treatment of ischaemic vascular diseases in clinical trials. We tested the hypothesis that mitochondrial depolarization stimulates the reparative functions of CD34+ cells. EXPERIMENTAL APPROACH Peripheral blood was obtained from healthy individuals (n = 63), and mononuclear cells (MNCs) were separated. MNCs were enriched for lineage negative cells, followed by isolation of CD34+ cells. Vascular repair-relevant functions of CD34+ cells, proliferation and migration, were evaluated in the presence and absence of diazoxide. Mitochondrial membrane potential, ROS and NO levels were evaluated by flow cytometry by using JC-1, mitoSOX and DAF-FM respectively. KEY RESULTS Diazoxide stimulated the proliferation and migration of CD34+ cells that were comparable to the responses induced by stromal-derived factor-1α (SDF) or VEGF. Effects of diazoxide were blocked by either 5-hydroxydecanoate (5HD), a selective mitochondrial ATP-sensitive potassium channel (mitoKATP ) inhibitor, or by L-NAME. Diazoxide induced mitochondrial depolarization, and NO and cGMP generation that were 5HD-sensitive. The generation of NO and cGMP by diazoxide was blocked by an endothelial NOS (eNOS)-selective inhibitor, NIO, but not by a neuronal (n)NOS-selective inhibitor, Nω -propyl-L-arginine (NPA). A Ca2+ chelator, BAPTA, Akt inhibitor, triciribine, or PI3K inhibitor, LY294002, inhibited the NO release induced by diazoxide. Phosphorylation of eNOS at Ser1177 and dephosphorylation at Thr495 were increased. Diazoxide-induced ROS generation and phosphorylation of eNOS at Ser1177 were reduced by NPA. CONCLUSION AND IMPLICATIONS Diazoxide stimulates vascular repair-relevant functions of CD34+ cells via the mitoKATP -dependent release of NO and ROS. LINKED ARTICLES This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Shrinidh Joshi
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Yagna P R Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| |
Collapse
|
14
|
|
15
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Tavares JGP, Errante PR, Govato TCP, Vasques ÊR, Ferraz RRN, Taha MO, Menezes-Rodrigues FS, Caricati-Neto A. Cardioprotective effect of preconditioning is more efficient than postconditioning in rats submitted to cardiac ischemia and reperfusion1. Acta Cir Bras 2018; 33:588-596. [PMID: 30110060 DOI: 10.1590/s0102-865020180070000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/24/2018] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To investigate the cardioprotective effects of ischemic preconditioning (preIC) and postconditioning (postIC) in animal model of cardiac ischemia/reperfusion. METHODS Adult rats were submitted to protocol of cardiac ischemia/reperfusion (I/R) and randomized into three experimental groups: cardiac I/R (n=33), preCI + cardiac I/R (n=7) and postCI + cardiac I/R (n=8). After this I/R protocol, the incidence of ventricular arrhythmia (VA), atrioventricular block (AVB) and lethality (LET) was evaluated using the electrocardiogram (ECG) analysis. RESULTS After reestablishment of coronary blood flow, we observed variations of the ECG trace with increased incidence of ventricular arrhythmia (VA) (85%), atrioventricular block (AVB) (79%), and increase of lethality (70%) in cardiac I/R group. The comparison between I/R + preIC group with I/R group demonstrated significant reduction in VA incidence to 28%, AVB to 0% and lethality to 14%. The comparison of I/R + postIC group with I/R group was observed significance reduction in AVB incidence to 25% and lethality to 25%. CONCLUSION The preconditioning strategies produce cardioprotection more efficient that postconditioning against myocardial dysfunctions and lethality by cardiac ischemia and reperfusion.
Collapse
Affiliation(s)
- José Gustavo Padrão Tavares
- Fellow PhD degree, Postgraduate Program in Pharmacology, Universidade Federal de São Paulo (UNIFESP), Brazil. Conception and design of the study, analysis and interpretation of data, manuscript writing
| | - Paolo Ruggero Errante
- Fellow PhD degree, Postgraduate Program in Pharmacology, UNIFESP, Sao Paulo-SP, Brazil. Analysis and interpretation of data, manuscript writing
| | - Tânia Carmem Peñaranda Govato
- Assistant Professor, Department of Pharmacology, Faculdade de Medicina do ABC (FMABC), Santo Andre-SP, Brazil. Statistical analysis
| | - Ênio Rodrigues Vasques
- Fellow PhD degree, Department of Gastroenterology, Faculty of Medicine, Universidade de São Paulo (USP), Brazil. Interpretation of electrocardiogram
| | - Renato Ribeiro Nogueira Ferraz
- Full Professor, Program in Management of Health System (PMPA-GSS), Universidade Nove de Julho (UNINOVE), Sao Paulo-SP, Brazil. Critical revision
| | - Murched Omar Taha
- Associate Professor, Department of Surgery, UNIFESP, Sao Paulo-SP, Brazil. Technical procedures
| | | | - Afonso Caricati-Neto
- Associate Professor, Department of Pharmacology, UNIFESP, Sao Paulo-SP, Brazil. Conception and design of the study, critical revision
| |
Collapse
|
17
|
|
18
|
Szeto V, Chen NH, Sun HS, Feng ZP. The role of K ATP channels in cerebral ischemic stroke and diabetes. Acta Pharmacol Sin 2018; 39:683-694. [PMID: 29671418 PMCID: PMC5943906 DOI: 10.1038/aps.2018.10] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022]
Abstract
ATP-sensitive potassium (KATP) channels are ubiquitously expressed on the plasma membrane of cells in multiple organs, including the heart, pancreas and brain. KATP channels play important roles in controlling and regulating cellular functions in response to metabolic state, which are inhibited by ATP and activated by Mg-ADP, allowing the cell to couple cellular metabolic state (ATP/ADP ratio) to electrical activity of the cell membrane. KATP channels mediate insulin secretion in pancreatic islet beta cells, and controlling vascular tone. Under pathophysiological conditions, KATP channels play cytoprotective role in cardiac myocytes and neurons during ischemia and/or hypoxia. KATP channel is a hetero-octameric complex, consisting of four pore-forming Kir6.x and four regulatory sulfonylurea receptor SURx subunits. These subunits are differentially expressed in various cell types, thus determining the sensitivity of the cells to specific channel modifiers. Sulfonylurea class of antidiabetic drugs blocks KATP channels, which are neuroprotective in stroke, can be one of the high stoke risk factors for diabetic patients. In this review, we discussed the potential effects of KATP channel blockers when used under pathological conditions related to diabetics and cerebral ischemic stroke.
Collapse
Affiliation(s)
- Vivian Szeto
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Nai-hong Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hong-shuo Sun
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
- Surgery
- Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Zhong-ping Feng
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
19
|
Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: a continual challenge. Eur Heart J 2018; 38:774-784. [PMID: 27354052 DOI: 10.1093/eurheartj/ehw224] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022] Open
Abstract
The incidence of ST segment elevation myocardial infarction (STEMI) has decreased over the last two decades in developed countries, but mortality from STEMI despite widespread access to reperfusion therapy is still substantial as is the development of heart failure, particularly among an expanding older population. In developing countries, the incidence of STEMI is increasing and interventional reperfusion is often not available. We here review the pathophysiology of acute myocardial infarction and reperfusion, notably the temporal and spatial evolution of ischaemic and reperfusion injury, the different modes of cell death, and the resulting coronary microvascular dysfunction. We then go on to briefly characterize the cardioprotective phenomena of ischaemic preconditioning, ischaemic postconditioning, and remote ischaemic conditioning and their underlying signal transduction pathways. We discuss in detail the attempts to translate conditioning strategies and drug therapy into the clinical setting. Most attempts have failed so far to reduce infarct size and improve clinical outcomes in STEMI patients, and we discuss potential reasons for such failure. Currently, it appears that remote ischaemic conditioning and a few drugs (atrial natriuretic peptide, exenatide, metoprolol, and esmolol) reduce infarct size, but studies with clinical outcome as primary endpoint are still underway.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Hufelandstr. 55, 45122 Essen, Germany
| | - Bernard J Gersh
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
20
|
Lee KY, Kim JR, Choi HC. Gliclazide, a K ATP channel blocker, inhibits vascular smooth muscle cell proliferation through the CaMKKβ-AMPK pathway. Vascul Pharmacol 2018; 102:21-28. [PMID: 29337033 DOI: 10.1016/j.vph.2018.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/31/2017] [Accepted: 01/10/2018] [Indexed: 10/18/2022]
Abstract
Gliclazide, a sulfonylurea that is widely used to treat type II-diabetes, specifically blocks KATP channels and recombinant smooth muscle (SUR2B/Kir6.1) KATP channels with high potency. Furthermore, it exerts antioxidant properties and inhibits tumor cell proliferation. In this study, we investigated the inhibitory effect of gliclazide on vascular smooth muscle cell (VSMC) proliferation and tried to identify the underlying signaling pathway. We first investigated the effect of gliclazide-induced AMP-activated protein kinase (AMPK) activation on the proliferation of VSMCs. Gliclazide induced phosphorylation of AMPK in a dose- and time-dependent manner and inhibited VSMC proliferation following stimulation by platelet-derived growth factor (PDGF). However, KATP channel openers and Kir6.1 siRNA prevented gliclazide-mediated inhibition of VSMC proliferation. Gliclazide also increased the levels of Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ), an upstream kinase of AMPK. These findings suggested that the effects of KATP channels on AMPK activity were mediated by the regulation of intracellular Ca2+ levels. Oral administration of 2mg/kg gliclazide resulted in the activation of CaMKKβ and AMPK in vivo, suggesting that gliclazide suppressed VSMC proliferation via the CaMKKβ-AMPK signaling pathway. Taken together, our observations indicated that gliclazide-induced AMPK activation may act to prevent diabetes-associated atherosclerosis.
Collapse
Affiliation(s)
- Kyung Young Lee
- Department of Pharmacology, Yeungnam University, 170 Hyunchung-Ro, Daegu 705-717, Republic of Korea; Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Daegu 705-717, Republic of Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University, 170 Hyunchung-Ro, Daegu 705-717, Republic of Korea; Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Daegu 705-717, Republic of Korea
| | - Hyoung Chul Choi
- Department of Pharmacology, Yeungnam University, 170 Hyunchung-Ro, Daegu 705-717, Republic of Korea; Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Daegu 705-717, Republic of Korea.
| |
Collapse
|
21
|
Connexin 43 and ATP-sensitive potassium channels crosstalk: a missing link in hypoxia/ischemia stress. Cell Tissue Res 2017; 371:213-222. [PMID: 29185069 DOI: 10.1007/s00441-017-2736-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/06/2017] [Indexed: 12/28/2022]
Abstract
Connexin 43 (Cx43) is a gap junction protein expressed in various tissues and organs of vertebrates. Besides functioning as a gap junction, Cx43 also regulates diverse cellular processes like cell growth and differentiation, cell migration, cell survival, etc. Cx43 is critical for normal cardiac functioning and is therefore abundantly expressed in cardiomyocytes. On the other hand, ATP-sensitive potassium (KATP) channels are metabolic sensors converting metabolic changes into electrical activity. These channels are important in maintaining the neurotransmitter release, smooth muscle relaxation, cardiac action potential repolarization, normal physiology of cellular repolarization, insulin secretion and immune function. Cx43 and KATP channels are part of the same signaling pathway, regulating cell survival during stress conditions and ischemia/hypoxia preconditioning. However, the underlying molecular mechanism for their combined role in ischemia/hypoxia preconditioning is largely unknown. The current review focuses on understanding the molecular mechanism responsible for the coordinated role of Cx43 and KATP channel protein in protecting cardiomyocytes against ischemia/hypoxia stress.
Collapse
|
22
|
Djafarzadeh S, Vuda M, Jeger V, Takala J, Jakob SM. The Effects of Fentanyl on Hepatic Mitochondrial Function. Anesth Analg 2017; 123:311-25. [PMID: 27089001 DOI: 10.1213/ane.0000000000001280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Remifentanil interferes with hepatic mitochondrial function. The aim of the present study was to evaluate whether hepatic mitochondrial function is affected by fentanyl, a more widely used opioid than remifentanil. METHODS Human hepatoma HepG2 cells were exposed to fentanyl or pretreated with naloxone (an opioid receptor antagonist) or 5-hydroxydecanoate (5-HD, an inhibitor of mitochondrial adenosine triphosphate (ATP)-sensitive potassium [mitoKATP] channels), followed by incubation with fentanyl. Mitochondrial function and metabolism were then analyzed. RESULTS Fentanyl marginally reduced maximal mitochondrial complex-specific respiration rates using exogenous substrates (decrease in medians: 11%-18%; P = 0.003-0.001) but did not affect basal cellular respiration rates (P = 0.834). The effect on stimulated respiration was prevented by preincubation with naloxone or 5-HD. Fentanyl reduced cellular ATP content in a dose-dependent manner (P < 0.001), an effect that was not significantly prevented by 5-HD and not explained by increased total ATPase concentration. However, in vitro ATPase activity of recombinant human permeability glycoprotein (an ATP-dependent drug efflux transporter) was significantly stimulated by fentanyl (P = 0.004). CONCLUSIONS Our data suggest that fentanyl reduces stimulated mitochondrial respiration of cultured human hepatocytes by a mechanism that is blocked by a mitoKATP channel antagonist. Increased energy requirements for fentanyl efflux transport may offer an explanation for the substantial decrease in cellular ATP concentration.
Collapse
Affiliation(s)
- Siamak Djafarzadeh
- From the *Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; and †Department of Clinical Research, Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | | | | | | |
Collapse
|
23
|
The Slo(w) path to identifying the mitochondrial channels responsible for ischemic protection. Biochem J 2017; 474:2067-2094. [PMID: 28600454 DOI: 10.1042/bcj20160623] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria play an important role in tissue ischemia and reperfusion (IR) injury, with energetic failure and the opening of the mitochondrial permeability transition pore being the major causes of IR-induced cell death. Thus, mitochondria are an appropriate focus for strategies to protect against IR injury. Two widely studied paradigms of IR protection, particularly in the field of cardiac IR, are ischemic preconditioning (IPC) and volatile anesthetic preconditioning (APC). While the molecular mechanisms recruited by these protective paradigms are not fully elucidated, a commonality is the involvement of mitochondrial K+ channel opening. In the case of IPC, research has focused on a mitochondrial ATP-sensitive K+ channel (mitoKATP), but, despite recent progress, the molecular identity of this channel remains a subject of contention. In the case of APC, early research suggested the existence of a mitochondrial large-conductance K+ (BK, big conductance of potassium) channel encoded by the Kcnma1 gene, although more recent work has shown that the channel that underlies APC is in fact encoded by Kcnt2 In this review, we discuss both the pharmacologic and genetic evidence for the existence and identity of mitochondrial K+ channels, and the role of these channels both in IR protection and in regulating normal mitochondrial function.
Collapse
|
24
|
Opening of the Adenosine Triphosphate-sensitive Potassium Channel Attenuates Morphine Tolerance by Inhibiting JNK and Astrocyte Activation in the Spinal Cord. Clin J Pain 2017; 32:617-23. [PMID: 26626290 DOI: 10.1097/ajp.0000000000000299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES In the present study, we investigated the role of adenosine triphosphate (ATP)-sensitive potassium (KATP) channels in chronic morphine tolerance. MATERIALS AND METHODS Male mice were injected intrathecally with morphine or saline, respectively (each in 10 μL). Different doses of the KATP opener cromakalim (0.3, 1, or 3 μg/10 μL/mouse) were administered 15 minutes before the morphine (10 μg/10 μL/mouse) challenge daily for 7 consecutive days. Half an hour after morphine injection, the tail-flick latency was measured to evaluate the antinociceptive effect of morphine. On the seventh day, mice were euthanized with sodium pentobarbital (100 mg/kg) at 1 hour after morphine injection, and their spinal cords were removed for the assays of Western blot, immunofluorescence, and quantitative real-time polymerase chain reaction. RESULTS Opening of the KATP channel attenuates chronic morphine tolerance, suppresses astrocyte activation inhibits the increase in interleukin-1β at the transcriptional and the translational levels, and reduces the upregulation of phosphorylated c-Jun N-terminal kinase mitogen-activated protein kinase in the spinal cord after chronic morphine treatment. Moreover, transcriptional levels of spinal cord astrocyte KATP channel subunits, named the inwardly rectifying potassium (Kir) 6.1 and sulfonylurea receptor 1, are decreased in morphine-tolerant mice. DISCUSSION Cromakalim suppresses morphine-induced astrocyte activation significantly by suppressing the c-Jun N-terminal kinase pathway, resulting in a reduced release of interleukin-1β and the attenuation of morphine chronic antinociceptive tolerance.
Collapse
|
25
|
Vornanen M. Electrical Excitability of the Fish Heart and Its Autonomic Regulation. FISH PHYSIOLOGY 2017. [DOI: 10.1016/bs.fp.2017.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
26
|
Zhang L, Cao S, Deng S, Yao G, Yu T. Ischemic postconditioning and pinacidil suppress calcium overload in anoxia-reoxygenation cardiomyocytes via down-regulation of the calcium-sensing receptor. PeerJ 2016; 4:e2612. [PMID: 27833799 PMCID: PMC5101590 DOI: 10.7717/peerj.2612] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/26/2016] [Indexed: 01/23/2023] Open
Abstract
Ischemic postconditioning (IPC) and ATP sensitive potassium channel (KATP) agonists (e.g. pinacidil and diazoxide) postconditioning are effective methods to defeat myocardial ischemia-reperfusion (I/R) injury, but their specific mechanisms of reducing I/R injury are not fully understood. We observed an intracellular free calcium ([Ca2+]i) overload in Anoxia/reoxygenation (A/R) cardiomyocytes, which can be reversed by KATP agonists diazoxide or pinacidil. The calcium-sensing receptor (CaSR) regulates intracellular calcium homeostasis. CaSR was reported to be involved in the I/R-induced apoptosis in rat cardiomyocytes. We therefore hypothesize that IPC and pinacidil postconditioning (PPC) reduce calcium overload in I/R cardiomyocytes by the down-regulation of CaSR. A/R model was established with adult rat caridomyocyte. mRNA and protein expression of CaSR were detected, IPC, PPC and KATP’s effects on [Ca2+]i concentration was assayed too. IPC and PPC ameliorated A/R insult induced [Ca2+]i overload in cardiomyocytes. In addition, they down-regulated the mRNA and protein level of CaSR as we expected. CaSR agonist spermine and KATP blocker glibenclamide offset IPC’s effects on CaSR expression and [Ca2+]i modulation. Our data indicate that CaSR down-regulation contributes to the mitigation of calcium overload in A/R cardiomyocytes, which may partially represents IPC and KATP’s myocardial protective mechanism under I/R circumstances.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Anesthesiology, Zunyi Medical College, Zunyi, Guizhou, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, Guizhou, China
| | - Song Cao
- Department of Anesthesiology, Zunyi Medical College, Zunyi, Guizhou, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, Guizhou, China
| | - Shengli Deng
- Department of Anesthesiology, Zunyi Medical College, Zunyi, Guizhou, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, Guizhou, China
| | - Gang Yao
- Department of Anesthesiology, Zunyi Medical College, Zunyi, Guizhou, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, Guizhou, China
| | - Tian Yu
- Department of Anesthesiology, Zunyi Medical College, Zunyi, Guizhou, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, Guizhou, China
| |
Collapse
|
27
|
Koo EH, Park YC, Lim SH, Kim HZ. Amiodarone Offsets the Cardioprotective Effects of Ischaemic Preconditioning against Ischaemia/Reperfusion Injury. J Int Med Res 2016; 34:140-51. [PMID: 16749409 DOI: 10.1177/147323000603400203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Both ischaemic preconditioning (IPC) and amiodarone protect against myocardial ischaemia. We examined whether a combination of IPC and amiodarone demonstrated an additive protective effect in isolated rat hearts ( n = 40). The controls (group I) were subjected to ischaemia/reperfusion injury; group II was subjected to cycles of IPC prior to ischaemia/reperfusion injury; group III was subjected to ischaemia in the presence of amiodarone (10−10 mol/l); and group IV was subjected to IPC followed by ischaemia in the presence of amiodarone (10−10 mol/l). Amiodarone produced the best preserved left ventricular end-systolic pressure and dP/dtmax, less developed ventricular stiffness, the shortest arrhythmia duration, and the smallest infarct size among the groups. All of the myocardial protective effects against ischaemia/reperfusion injury were diminished or abolished when IPC and amiodarone were applied sequentially.
Collapse
Affiliation(s)
- E H Koo
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Guro Hospital, Seoul, South Korea
| | | | | | | |
Collapse
|
28
|
Abstract
Multiple types of voltage-gated K(+) and non-voltage-gated K(+) currents have been distinguished in mammalian cardiac myocytes based on differences in time-dependent and voltage-dependent properties and pharmacologic sensitivities. Many of the genes encoding voltage-gated K(+) (Kv) and non-voltage-gated K(+) (Kir and K2P) channel pore-forming and accessory subunits are expressed in the heart, and a variety of approaches have been, and continue to be, used to define the molecular determinants of native cardiac K(+) channels and to explore the molecular mechanisms controlling the diversity, regulation, and remodeling of these channels in the normal and diseased myocardium.
Collapse
Affiliation(s)
- Jeanne M Nerbonne
- Department of Internal Medicine, Washington University Medical School, 660 South Euclid Avenue, Box 8086, St Louis, MO 63110, USA; Department of Developmental Biology, Washington University Medical School, St Louis, MO 63110, USA.
| |
Collapse
|
29
|
Shimizu Y, Calvert JW. Recycling KATP channels for cardioprotection. Am J Physiol Heart Circ Physiol 2016; 310:H1381-2. [PMID: 27199122 DOI: 10.1152/ajpheart.00309.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuuki Shimizu
- Carlyle Fraser Heart Center of Emory University Hospital Midtown, Atlanta, Georgia; and Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta Georgia
| | - John W Calvert
- Carlyle Fraser Heart Center of Emory University Hospital Midtown, Atlanta, Georgia; and Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, Atlanta Georgia
| |
Collapse
|
30
|
Tomson TT, Arora R. Modulation of Cardiac Potassium Current by Neural Tone and Ischemia. Card Electrophysiol Clin 2016; 8:349-60. [PMID: 27261826 DOI: 10.1016/j.ccep.2016.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cardiac action potential is generated by intricate flows of ions across myocyte cell membranes in a coordinated fashion to control myocardial contraction and the heart rhythm. Modulation of the flow of these ions in response to a variety of stimuli results in changes to the action potential. Abnormal or altered ion currents can result in cardiac arrhythmias. Abnormalities of autonomic regulation of potassium current play a role in the genesis of cardiac arrhythmias, and alterations in acetylcholine-activated potassium channels may play a key role in atrial fibrillation. Ischemia is another important modulator of cardiac cellular electrophysiology.
Collapse
Affiliation(s)
- Todd T Tomson
- Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rishi Arora
- Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
31
|
Goharbari MH, Shadboores A, Abdollahi M. Inhibitory Effects of Thyroid Hormones on Mitochondrial
Oxidative Stress: A Systematic Review. INT J PHARMACOL 2016. [DOI: 10.3923/ijp.2016.249.261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Jameel MN, Xiong Q, Mansoor A, Bache RJ, Zhang J. ATP sensitive K(+) channels are critical for maintaining myocardial perfusion and high energy phosphates in the failing heart. J Mol Cell Cardiol 2016; 92:116-21. [PMID: 26854629 DOI: 10.1016/j.yjmcc.2016.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/21/2016] [Accepted: 02/04/2016] [Indexed: 01/08/2023]
Abstract
Congestive heart failure (CHF) is associated with intrinsic alterations of mitochondrial oxidative phosphorylation which lead to increased myocardial cytosolic free ADP. ATP sensitive K(+) channels (KATP) act as metabolic sensors that are important for maintaining coronary blood flow (MBF) and in mediating the response of the myocardium to stress. Coronary adenosine receptors (AdR) are not normally active but cause vasodilation during myocardial ischemia. This study examined the myocardial energetic response to inhibition of KATP and AdR in CHF. CHF (as evidenced by LVEDP>20mmHg) was produced in adult mongrel dogs (n=12) by rapid ventricular pacing for 4weeks. MBF was measured with radiolabeled microspheres during baseline (BL), AdR blockade with 8-phenyltheophylline (8-PT; 5mg/kg iv), and KATP blockade with glibenclamide (GLB; 20μg/kg/min ic). High energy phosphates were examined with (31)P magnetic resonance spectroscopy (MRS) while myocardial oxygenation was assessed from the deoxymyoglobin signal (Mb-δ) using (1)H MRS. During basal conditions the phosphocreatine (PCr)/ATP ratio (1.73±0.15) was significantly lower than in previously studied normal dogs (2.42±0.11) although Mb-δ was undetectable. 8-PT caused ≈21% increase in MBF with no change in PCr/ATP. GLB caused a 33±0.1% decrease in MBF with a decrease in PCr/ATP from 1.65±0.17 to 1.11±0.11 (p<0.0001). GLB did not change the pseudo-first-order rate constant of ATP production via CK (kf), but the ATP production rate via CK was reduced by 35±0.08%; this was accompanied by an increase in Pi/PCr and appearance of a Mb-δ signal indicating tissue hypoxia. Thus, in the failing heart the balance between myocardial ATP demands and oxygen delivery is critically dependent on functioning KATP channels.
Collapse
Affiliation(s)
- Mohammad N Jameel
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Qiang Xiong
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Abdul Mansoor
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Robert J Bache
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jianyi Zhang
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
33
|
Ion Channels and Oxidative Stress as a Potential Link for the Diagnosis or Treatment of Liver Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3928714. [PMID: 26881024 PMCID: PMC4736365 DOI: 10.1155/2016/3928714] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/22/2015] [Accepted: 10/27/2015] [Indexed: 02/06/2023]
Abstract
Oxidative stress results from a disturbed balance between oxidation and antioxidant systems. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) may be either harmful or beneficial to the cells. Ion channels are transmembrane proteins that participate in a large variety of cellular functions and have been implicated in the development of a variety of diseases. A significant amount of the available drugs in the market targets ion channels. These proteins have sulfhydryl groups of cysteine and methionine residues in their structure that can be targeted by ROS and RNS altering channel function including gating and conducting properties, as well as the corresponding signaling pathways associated. The regulation of ion channels by ROS has been suggested to be associated with some pathological conditions including liver diseases. This review focuses on understanding the role and the potential association of ion channels and oxidative stress in liver diseases including fibrosis, alcoholic liver disease, and cancer. The potential association between ion channels and oxidative stress conditions could be used to develop new treatments for major liver diseases.
Collapse
|
34
|
Liu Z, Cai H, Dang Y, Qiu C, Wang J. Adenosine triphosphate-sensitive potassium channels and cardiomyopathies (Review). Mol Med Rep 2015; 13:1447-54. [PMID: 26707080 DOI: 10.3892/mmr.2015.4714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/05/2015] [Indexed: 11/06/2022] Open
Abstract
Cardiomyopathies have been indicated to be one of the leading causes of heart failure. Though it was indicated that genetic defects, viral infection and trace element deficiency were among the causes of cardiomyopathy, the etiology has remained to be fully elucidated. Cardiomyocytes require large amounts of energy to maintain their normal biological functions. Adenosine triphosphate-sensitive potassium channels (KATP), composed of inward-rectifier potassium ion channel and sulfonylurea receptor subunits, are present on the cell surface and mitochondrial membrane of cardiac muscle cells. As metabolic sensors sensitive to changes in intracellular energy levels, KATP adapt electrical activities to metabolic challenges, maintaining normal biological functions of myocytes. It is implied that malfunctions, mutations and altered expression of KATP are associated with the pathogenesis of conditions including c hypertrophy, diabetes as well as dilated, ischemic and endemic cardiomyopathy. However, the current knowledge is only the tip of the iceberg and the roles of KATP in cardiomyopathies largely remain to be elucidated in future studies.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Hui Cai
- Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yonghui Dang
- College of Medicine and Forensics, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Chuan Qiu
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans 70112‑2705, LA, USA
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
35
|
Van Caenegem O, Beauloye C, Bertrand L, Horman S, Lepropre S, Sparavier G, Vercruysse J, Bethuyne N, Poncelet AJ, Gianello P, Demuylder P, Legrand E, Beaurin G, Bontemps F, Jacquet LM, Vanoverschelde JL. Hypothermic continuous machine perfusion enables preservation of energy charge and functional recovery of heart grafts in an ex vivo model of donation following circulatory death. Eur J Cardiothorac Surg 2015; 49:1348-53. [PMID: 26604296 DOI: 10.1093/ejcts/ezv409] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/13/2015] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Cardiac transplantation using hearts from donors after circulatory death (DCD) is critically limited by the unavoidable warm ischaemia and its related unpredictable graft function. Inasmuch as hypothermic machine perfusion (MP) has been shown to improve heart preservation, we hypothesized that MP could enable the use of DCD hearts for transplantation. METHODS We recovered 16 pig hearts following anoxia-induced cardiac arrest and cardioplegia. Grafts were randomly assigned to two different groups of 4-h preservation using either static cold storage (CS) or MP (Modified LifePort© System, Organ Recovery Systems©, Itasca, Il). After preservation, the grafts were reperfused ex vivo using the Langendorff method for 60 min. Energetic charge was quantified at baseline, post-preservation and post-reperfusion by measuring lactate and high-energy phosphate levels. Left ventricular contractility parameters were assessed both in vivo prior to ischaemia and ex vivo during reperfusion. RESULTS Following preservation, the hearts that were preserved using CS exhibited higher lactate levels (57.1 ± 23.7 vs 21.4 ± 12.2 µmol/g; P < 0.001), increased adenosine monophosphate/adenosine triphosphate ratio (0.53 ± 0.25 vs 0.11 ± 0.11; P < 0.001) and lower phosphocreatine/creatine ratio (9.7 ± 5.3 vs 25.2 ± 11; P < 0.001) in comparison with the MP hearts. Coronary flow was similar in both groups during reperfusion (107 ± 9 vs 125 ± 9 ml/100 g/min heart; P = ns). Contractility decreased in the CS group, yet remained well preserved in the MP group. CONCLUSION MP preservation of DCD hearts results in improved preservation of the energy and improved functional recovery of heart grafts compared with CS.
Collapse
Affiliation(s)
- Olivier Van Caenegem
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium Cardiovascular Intensive Care, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Christophe Beauloye
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium Cardiovascular Intensive Care, Cliniques universitaires Saint Luc, Brussels, Belgium Division of Cardiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Sandrine Horman
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Sophie Lepropre
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Grégory Sparavier
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | | | - Noëlla Bethuyne
- Division of Cardiac Surgery, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Alain J Poncelet
- Division of Cardiac Surgery, Cliniques universitaires Saint Luc, Brussels, Belgium Pôle de chirurgie expérimentale et transplantation, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Pierre Gianello
- Pôle de chirurgie expérimentale et transplantation, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | | | - Eric Legrand
- Pôle de chirurgie expérimentale et transplantation, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Gwen Beaurin
- Pôle de chirurgie expérimentale et transplantation, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Bontemps
- Pôle de biochimie et recherche métabolique, Institut de Duve, Université catholique de Louvain, Brussels, Belgium
| | - Luc M Jacquet
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium Cardiovascular Intensive Care, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Jean-Louis Vanoverschelde
- Pôle de recherche cardiovasculaire, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium Division of Cardiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
36
|
Bayat G, Javan M, Safari F, Khalili A, Shokri S, Goudarzvand M, Salimi M, Hajizadeh S. Nandrolone decanoate negatively reverses the beneficial effects of exercise on cardiac muscle via sarcolemmal, but not mitochondrial K(ATP) channel. Can J Physiol Pharmacol 2015; 94:324-31. [PMID: 26909616 DOI: 10.1139/cjpp-2015-0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
ATP-sensitive potassium channels are supposed to have a substantial role in improvement of cardiac performance. This study was performed to evaluate whether nandrolone decanoate (ND) and (or) exercise training could affect the expression of cardiac K(ATP) channel subunits. Thirty-five male albino Wistar rats were randomly divided into 5 groups, including sedentary control (SC), sedentary vehicle (SV), sedentary ND (SND), exercise control (EC), and exercise and ND (E+ND). Exercise training was performed on a treadmill 5 times per week. ND was injected (10 mg/kg/week, i.m.) to the rats in the SND and E+ND groups. Following cardiac isolation, the expression of both sarcolemmal and mitochondrial subunits of K(ATP) channel was measured using Western blot method. The expression of sarcolemmal, but not mitochondrial, subunits of K(ATP) channel (Kir6.2 and SUR2) of EC group was significantly higher compared with SC group while ND administration (SND group) did not show any change in their expression. In the E+ND group, ND administration led to decrease of the over-expression of sarcolemmal Kir6.2 and SUR2 which was previously induced by exercise. There was no significant association between the mitochondrial expression of either Kir6.2 or SUR2 proteins and administration of ND or exercise. Supra-physiological dosage of ND negatively reverses the effects of exercise on the cardiac muscle expression of sarcolemmal, but not mitochondrial, K(ATP) channel subunits.
Collapse
Affiliation(s)
- Gholamreza Bayat
- a Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Javan
- b Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Safari
- c Deptartment of Physiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azadeh Khalili
- d Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Shokri
- e Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdi Goudarzvand
- a Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehdi Salimi
- b Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sohrab Hajizadeh
- b Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
37
|
Kostic J, Djordjevic-Dikic A, Dobric M, Milasinovic D, Nedeljkovic M, Stojkovic S, Stepanovic J, Tesic M, Trifunovic Z, Zamaklar-Tifunovic D, Radosavljevic-Radovanovic M, Ostojic M, Beleslin B. The effects of nicorandil on microvascular function in patients with ST segment elevation myocardial infarction undergoing primary PCI. Cardiovasc Ultrasound 2015; 13:26. [PMID: 26012474 PMCID: PMC4446834 DOI: 10.1186/s12947-015-0020-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/18/2015] [Indexed: 11/10/2022] Open
Abstract
Background Nicorandil, as a selective potassium channel opener, has dual action including coronary and peripheral vasodilatation and cardioprotective effect through ischemic preconditioning. Considering those characteristics, nicorandil was suggested to reduce the degree of microvascular dysfunction. Methods Thirty-two patients with ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention (pPCI) were included in the study. Index of microvascular resistance (IMR) was measured in all patients immediatelly after pPCI before the after administration of Nicorandil. ST segment resolution was monitored before intervention and 60 min after terminating the procedure. Echocardiographic evaluation of myocardial function and transthoracic Doppler derived Coronary flow reserve (CFR) of infarct related artery (IRA) was performed during hospitalization and 3 months later. Results IMR was significantly lower after administration of Nicorandil (9.9 ± 3.7 vs. 14.1 ± 5.1, p < 0.001). There was significant difference in ST segment elevation before and after primary PCI with administration of Nicorandil (6.9 ± 3.7 mm vs. 1.6 ± 1.6 mm, p < 0.001). Transthoracic Doppler CFR measurement improved after 3 months (2.69 ± 0.38 vs. 2.92 ± 0.54, p = 0.021), as well as WMSI (1.14 ± 0.17 vs. 1.07 ± 0.09, p = 0.004). Conclusion Intracoronary Nicorandil administration after primary PCI significantly decreases IMR, resulting in improved CFR and ventricular function in patients with STEMI undergoing primary PCI.
Collapse
Affiliation(s)
- Jelena Kostic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia.
| | - Ana Djordjevic-Dikic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | - Milan Dobric
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | - Dejan Milasinovic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia.
| | - Milan Nedeljkovic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | - Sinisa Stojkovic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | - Jelena Stepanovic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | - Milorad Tesic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia.
| | - Zoran Trifunovic
- Military Medical Academy, Belgrade, Serbia. .,Medical Faculty, University of Defense, Belgrade, Serbia.
| | - Danijela Zamaklar-Tifunovic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | - Mina Radosavljevic-Radovanovic
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| | | | - Branko Beleslin
- Clinic for Cardiology, Clinical Center of Serbia, Visegradska 26, Belgrade, Serbia. .,Medical School, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
38
|
Liu X, Sun K, Song A, Zhang X, Zhang X, He X. Curcumin inhibits proliferation of gastric cancer cells by impairing ATP-sensitive potassium channel opening. World J Surg Oncol 2014; 12:389. [PMID: 25523120 PMCID: PMC4395964 DOI: 10.1186/1477-7819-12-389] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND This study was aimed to investigate whether ATP-sensitive potassium channel (KATP) is involved in curcumin's anti-proliferative effects against gastric cancer. METHODS In an in vitro study, gastric cancer cell line SGC-7901 was treated with curcumin at serial concentrations and co-administrated with the KATP opener, diazoxide. The effect of curcumin and diazoxide on proliferation were assessed by MTT assay. Mitochondrial membrane potential (MMP) was studied by flow cytometry detection of rhodamine 123 staining. Apoptosis was evaluated by flow cytometry detection of Annexin V propidium iodide double staining. In an in vivo study, SGC-7901 cells were planted into nude mice as xenografts. Animals were treated with curcumin co-administered with diazoxide. Tumor volume and tumor weight were observed. RESULTS Curcumin incubation significantly induced loss of MMP in SGC-7901 cells in a dose- dependent manner (P < 0.05); the cell apoptotic rate also dramatically increased after curcumin incubation in a dose-dependent manner (P < 0.05). After co-administration with diazoxide, however, we found that both the MMP-loss-inducing and the apoptosis-inducing effects of curcumin in SGC-7901 cells were significantly impaired (all P < 0.05). As a result, the proliferation of SGC-7901 cells was maintained by diazoxide treatment. CONCLUSIONS Impaired mitoKATP opening causes MMP loss, and is involved in curcumin-induced apoptosis in gastric cancer.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of General Surgery, second affiliated hospital of Lanzhou University, Lanzhou, Gansu, 730000, China.
| | - Kai Sun
- The Second Department of Thoracic Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Ailin Song
- Department of General Surgery, second affiliated hospital of Lanzhou University, Lanzhou, Gansu, 730000, China.
| | - Xiaoyun Zhang
- Department of Pathology, Lanzhou University Medical School, 199 West Donggang Road, Lanzhou, Gansu, 730000, China.
| | - Xu Zhang
- Department of Pathology, Lanzhou University Medical School, 199 West Donggang Road, Lanzhou, Gansu, 730000, China.
| | - Xiaodong He
- Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
39
|
Abstract
INTRODUCTION Cardiac K(+) channels play a critical role in maintaining the normal electrical activity of the heart by setting the cell resting membrane potential and by determining the shape and duration of the action potential. Drugs that block the rapid (IKr) and slow (IKs) components of the delayed rectifier K(+) current have been widely used as class III antiarrhythmic agents. In addition, drugs that selectively target the ultra-rapid delayed rectifier current (IKur) and the acetylcholine-gated inward rectifier current (IKAch) have shown efficacy in the treatment of patients with atrial fibrillation. In order to meet the future demand for new antiarrhythmic agents, novel approaches for cardiac K(+) channel drug discovery will need to be developed. Further, K(+) channel screening assays utilizing primary and stem cell-derived cardiomyocytes will be essential for evaluating the cardiotoxicity of potential drug candidates. AREAS COVERED In this review, the author provides a brief background on the structure, function and pharmacology of cardiac voltage-gated and inward rectifier K(+) channels. He then focuses on describing and evaluating current technologies, such as ion flux and membrane potential-sensitive dye assays, used for cardiac K(+) channel drug discovery. EXPERT OPINION Cardiac K(+) channels will continue to represent significant clinical targets for drug discovery. Although fluorescent high-throughput screening (HTS) assays and automated patch clamp systems will remain the workhorse technologies for identifying lead compounds, innovations in the areas of microfluidics, micropatterning and biosensor fabrication will allow further growth of technologies using primary and stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Kenneth B Walsh
- University of South Carolina, School of Medicine, Department of Pharmacology, Physiology and Neuroscience , Columbia, SC 29209 , USA +1 803 216 3519 ; +1 803 216 3538 ;
| |
Collapse
|
40
|
Brennan S, Jackson R, Patel M, Sims MW, Hudman D, Norman RI, Lodwick D, Rainbow RD. Early opening of sarcolemmal ATP-sensitive potassium channels is not a key step in PKC-mediated cardioprotection. J Mol Cell Cardiol 2014; 79:42-53. [PMID: 25450614 DOI: 10.1016/j.yjmcc.2014.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 09/29/2014] [Accepted: 10/20/2014] [Indexed: 11/29/2022]
Abstract
ATP-sensitive potassium (KATP) channels are abundantly expressed in the myocardium. Although a definitive role for the channel remains elusive they have been implicated in the phenomenon of cardioprotection, but the precise mechanism is unclear. We set out to test the hypothesis that the channel protects by opening early during ischemia to shorten action potential duration and reduce electrical excitability thus sparing intracellular ATP. This could reduce reperfusion injury by improving calcium homeostasis. Using a combination of contractile function analysis, calcium fluorescence imaging and patch clamp electrophysiology in cardiomyocytes isolated from adult male Wistar rats, we demonstrated that the opening of sarcolemmal KATP channels was markedly delayed after cardioprotective treatments: ischemic preconditioning, adenosine and PMA. This was due to the preservation of intracellular ATP for longer during simulated ischemia therefore maintaining sarcolemmal KATP channels in the closed state for longer. As the simulated ischemia progressed, KATP channels opened to cause contractile, calcium transient and action potential failure; however there was no indication of any channel activity early during simulated ischemia to impart an energy sparing hyperpolarization or action potential shortening. We present compelling evidence to demonstrate that an early opening of sarcolemmal KATP channels during simulated ischemia is not part of the protective mechanism imparted by ischemic preconditioning or other PKC-dependent cardioprotective stimuli. On the contrary, channel opening was actually delayed. We conclude that sarcolemmal KATP channel opening is a consequence of ATP depletion, not a primary mechanism of ATP preservation in these cells.
Collapse
Affiliation(s)
- Sean Brennan
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK
| | - Robert Jackson
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK
| | - Manish Patel
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK
| | - Mark W Sims
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK
| | - Diane Hudman
- Department of Medical and Social Care Education, Maurice Shock Medical Sciences Building, University of Leicester, Leicester, LE1 9HN, UK
| | - Robert I Norman
- Department of Medical and Social Care Education, Maurice Shock Medical Sciences Building, University of Leicester, Leicester, LE1 9HN, UK
| | - David Lodwick
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK
| | - Richard D Rainbow
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, LE3 9QP, UK.
| |
Collapse
|
41
|
Tinker A, Aziz Q, Thomas A. The role of ATP-sensitive potassium channels in cellular function and protection in the cardiovascular system. Br J Pharmacol 2014; 171:12-23. [PMID: 24102106 DOI: 10.1111/bph.12407] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/30/2013] [Accepted: 08/26/2013] [Indexed: 12/14/2022] Open
Abstract
ATP-sensitive potassium channels (K(ATP)) are widely distributed and present in a number of tissues including muscle, pancreatic beta cells and the brain. Their activity is regulated by adenine nucleotides, characteristically being activated by falling ATP and rising ADP levels. Thus, they link cellular metabolism with membrane excitability. Recent studies using genetically modified mice and genomic studies in patients have implicated K(ATP) channels in a number of physiological and pathological processes. In this review, we focus on their role in cellular function and protection particularly in the cardiovascular system.
Collapse
Affiliation(s)
- Andrew Tinker
- William Harvey Heart Centre, Barts and The London School of Medicine and Dentistry, London, UK
| | | | | |
Collapse
|
42
|
Raphemot R, Swale DR, Dadi PK, Jacobson DA, Cooper P, Wojtovich AP, Banerjee S, Nichols CG, Denton JS. Direct activation of β-cell KATP channels with a novel xanthine derivative. Mol Pharmacol 2014; 85:858-65. [PMID: 24646456 PMCID: PMC4014665 DOI: 10.1124/mol.114.091884] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/19/2014] [Indexed: 11/22/2022] Open
Abstract
ATP-regulated potassium (KATP) channel complexes of inward rectifier potassium channel (Kir) 6.2 and sulfonylurea receptor (SUR) 1 critically regulate pancreatic islet β-cell membrane potential, calcium influx, and insulin secretion, and consequently, represent important drug targets for metabolic disorders of glucose homeostasis. The KATP channel opener diazoxide is used clinically to treat intractable hypoglycemia caused by excessive insulin secretion, but its use is limited by off-target effects due to lack of potency and selectivity. Some progress has been made in developing improved Kir6.2/SUR1 agonists from existing chemical scaffolds and compound screening, but there are surprisingly few distinct chemotypes that are specific for SUR1-containing KATP channels. Here we report the serendipitous discovery in a high-throughput screen of a novel activator of Kir6.2/SUR1: VU0071063 [7-(4-(tert-butyl)benzyl)-1,3-dimethyl-1H-purine-2,6(3H,7H)-dione]. The xanthine derivative rapidly and dose-dependently activates Kir6.2/SUR1 with a half-effective concentration (EC50) of approximately 7 μM, is more efficacious than diazoxide at low micromolar concentrations, directly activates the channel in excised membrane patches, and is selective for SUR1- over SUR2A-containing Kir6.1 or Kir6.2 channels, as well as Kir2.1, Kir2.2, Kir2.3, Kir3.1/3.2, and voltage-gated potassium channel 2.1. Finally, we show that VU0071063 activates native Kir6.2/SUR1 channels, thereby inhibiting glucose-stimulated calcium entry in isolated mouse pancreatic β cells. VU0071063 represents a novel tool/compound for investigating β-cell physiology, KATP channel gating, and a new chemical scaffold for developing improved activators with medicinal chemistry.
Collapse
Affiliation(s)
- Rene Raphemot
- Departments of Anesthesiology (R.R., D.R.S., S.B., J.S.D.), Pharmacology (R.R., J.S.D.), and Molecular Physiology and Biophysics (P.K.D., D.A.J.) and Institutes of Chemical Biology (J.S.D.) and Global Health (J.S.D.), Vanderbilt University Medical Center, Nashville, Tennessee; Department of Medicine, University of Rochester Medical Center, Rochester, New York (A.P.W.); and Department of Cell Biology and Physiology (P.C., C.G.N.) and Center for the Investigation of Membrane Excitability Disorders (P.C., C.G.N.), Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Label-free cell phenotypic profiling decodes the composition and signaling of an endogenous ATP-sensitive potassium channel. Sci Rep 2014; 4:4934. [PMID: 24816792 PMCID: PMC4017216 DOI: 10.1038/srep04934] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/24/2014] [Indexed: 11/27/2022] Open
Abstract
Current technologies for studying ion channels are fundamentally limited because of their inability to functionally link ion channel activity to cellular pathways. Herein, we report the use of label-free cell phenotypic profiling to decode the composition and signaling of an endogenous ATP-sensitive potassium ion channel (KATP) in HepG2C3A, a hepatocellular carcinoma cell line. Label-free cell phenotypic agonist profiling showed that pinacidil triggered characteristically similar dynamic mass redistribution (DMR) signals in A431, A549, HT29 and HepG2C3A, but not in HepG2 cells. Reverse transcriptase PCR, RNAi knockdown, and KATP blocker profiling showed that the pinacidil DMR is due to the activation of SUR2/Kir6.2 KATP channels in HepG2C3A cells. Kinase inhibition and RNAi knockdown showed that the pinacidil activated KATP channels trigger signaling through Rho kinase and Janus kinase-3, and cause actin remodeling. The results are the first demonstration of a label-free methodology to characterize the composition and signaling of an endogenous ATP-sensitive potassium ion channel.
Collapse
|
44
|
Xu A, Szczepanek K, Maceyka MW, Ross T, Bowler E, Hu Y, Kenny B, Mehfoud C, Desai PN, Baumgarten CM, Chen Q, Lesnefsky EJ. Transient complex I inhibition at the onset of reperfusion by extracellular acidification decreases cardiac injury. Am J Physiol Cell Physiol 2014; 306:C1142-53. [PMID: 24696146 DOI: 10.1152/ajpcell.00241.2013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A reversible inhibition of mitochondrial respiration by complex I inhibition at the onset of reperfusion decreases injury in buffer-perfused hearts. Administration of acidic reperfusate for a brief period at reperfusion decreases cardiac injury. We asked if acidification treatment decreased cardiac injury during reperfusion by inhibiting complex I. Exposure of isolated mouse heart mitochondria to acidic buffer decreased the complex I substrate-stimulated respiration, whereas respiration with complex II substrates was unaltered. Evidence of the rapid and reversible inhibition of complex I by an acidic environment was obtained at the level of isolated complex, intact mitochondria and in situ mitochondria in digitonin-permeabilized cardiac myocytes. Moreover, ischemia-damaged complex I was also reversibly inhibited by an acidic environment. In the buffer-perfused mouse heart, reperfusion with pH 6.6 buffer for the initial 5 min decreased infarction. Compared with untreated hearts, acidification treatment markedly decreased the mitochondrial generation of reactive oxygen species and improved mitochondrial calcium retention capacity and inner mitochondrial membrane integrity. The decrease in infarct size achieved by acidic reperfusion approximates the reduction obtained by a reversible, partial blockade of complex I at reperfusion. Extracellular acidification decreases cardiac injury during reperfusion in part via the transient and reversible inhibition of complex I, leading to a reduction of oxyradical generation accompanied by a decreased susceptibility to mitochondrial permeability transition during early reperfusion.
Collapse
Affiliation(s)
- Aijun Xu
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; and
| | - Karol Szczepanek
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Michael W Maceyka
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Thomas Ross
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Elizabeth Bowler
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; University of the West of England, Bristol, United Kingdom
| | - Ying Hu
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Barrett Kenny
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chris Mehfoud
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Pooja N Desai
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Clive M Baumgarten
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Qun Chen
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; McGuire Veterans Affairs Medical Center, Richmond, Virginia;
| |
Collapse
|
45
|
The mitochondria as a target for cardioprotection in acute myocardial ischemia. Pharmacol Ther 2014; 142:33-40. [DOI: 10.1016/j.pharmthera.2013.11.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/01/2013] [Indexed: 12/28/2022]
|
46
|
Abstract
A selective history of the pathophysiological, structural, and metabolic changes found during an episode of severe myocardial ischemia in the canine heart is presented. The changes that cause ischemic injury to become irreversible are discussed in detail because these changes are the target of any successful therapy designed to prevent ischemic cell death. Of these, the disruption of the sarcolemma, an injury the development of which is accelerated in vivo by the contraction of viable tissue elsewhere in the heart traumatizing the ischemic area, plus the changes in high-energy phosphate and the total adenine nucleotide pool are considered to be the critical events leading to the development of irreversibility. The discovery of preconditioning with ischemia is discussed, together with a brief description of postconditioning. Finally, reperfusion injury is discussed in a summary fashion. The evidence for the fact that myocytes are salvaged by reperfusion is presented, as is the evidence that myocytes become unsalvageable by reperfusion as the duration of ischemia increases. The concept that some of the myocytes that die after successful reperfusion with arterial blood actually are killed by changes initiated by reperfusion, so-called lethal reperfusion injury, is attractive in that prevention of this change would lead to greater salvage; however, the prevalence of this phenomenon in clinical practice remains to be determined.
Collapse
Affiliation(s)
- Robert B Jennings
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
47
|
Ischemic preconditioning protects cardiomyocyte mitochondria through mechanisms independent of cytosol. J Mol Cell Cardiol 2014; 68:79-88. [PMID: 24434643 DOI: 10.1016/j.yjmcc.2014.01.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/03/2014] [Indexed: 12/22/2022]
Abstract
Mitochondria play a central role in the protection conferred by ischemic preconditioning (IP) by not fully elucidated mechanisms. We investigated whether IP protects mitochondria against ischemia-reperfusion (IR) injury through mechanisms independent of cytosolic signaling. In isolated rat hearts, sublethal IR increased superoxide production and reduced complex-I- and II-mediated respiration in subsarcolemmal (SS), but not interfibrillar (IF) mitochondria. This effect of IR on mitochondrial respiration was significantly attenuated by IP. Similar results were obtained in isolated cardiac mitochondria subjected to in vitro IR. The reduction in SS mitochondrial respiration in the heart and in vitro model was paralleled by an increase in oxidized cysteine residues, which was also prevented by IP. IP was also protective in mitochondria submitted to lethal IR. The protective effect of IP against respiratory failure was unaffected by inhibition of mitochondrial KATP channels or mitochondrial permeability transition. However, IP protection was lost in mitochondria from genetically-modified animals in which connexin-43, a protein present in SS but not IF mitochondria, was replaced by connexin-32. Our results demonstrate the existence of a protective mitochondrial mechanism or "mitochondrial preconditioning" independent of cytosol that confers protection against IR-induced respiratory failure and oxidative damage, and requires connexin-43.
Collapse
|
48
|
Tian C, Zhu R, Zhu L, Qiu T, Cao Z, Kang T. Potassium Channels: Structures, Diseases, and Modulators. Chem Biol Drug Des 2013; 83:1-26. [DOI: 10.1111/cbdd.12237] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Chuan Tian
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| | - Ruixin Zhu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Lixin Zhu
- Department of Pediatrics; Digestive Diseases and Nutrition Center; The State University of New York at Buffalo; Buffalo NY 14226 USA
| | - Tianyi Qiu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Zhiwei Cao
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Tingguo Kang
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| |
Collapse
|
49
|
Venediktova N, Shigaeva M, Belova S, Belosludtsev K, Belosludtseva N, Gorbacheva O, Lezhnev E, Lukyanova L, Mironova G. Oxidative phosphorylation and ion transport in the mitochondria of two strains of rats varying in their resistance to stress and hypoxia. Mol Cell Biochem 2013; 383:261-269. [PMID: 23943284 DOI: 10.1007/s11010-013-1774-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/02/2013] [Indexed: 01/23/2023]
Abstract
The role of mitochondria in the inherited or ontogenetically acquired reactions of organism to stress is not studied enough. In the present work, we examined the functional state of the coupled respiratory chain, potassium and calcium transport and rate of hydrogen peroxide production on two rat lines: August and Wistar-which possess different resistance to emotional stress and hypoxia. It was established that the respiration rate and efficiency of oxidative phosphorylation were higher in August rats than in Wistar ones. In August rats, the rate of potassium transport and ATP-dependent mitochondrial swelling as well as the concentration of the ion in the mitochondrial matrix were almost twice as higher comparatively to those parameters in Wistar rats. The rate of H2O2 production was found to be decreased in the mitochondria of August rats. It was also demonstrated that the two rat lines differed by their resistance to the opening of the palmitate/Ca(2+)-induced pore and by their ability to retain calcium within mitochondria. The paper discusses the involvement of the mitochondrial ATP-dependent potassium channel in the adaptation of animals to adverse effects.
Collapse
Affiliation(s)
- N Venediktova
- Institute of Theoretical and Experimental Biophysics (RAS), Pushchino, Moscow Region, 142290, Russia,
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Murzaeva SV, Belova SP, Mironova GD. Determination of the antioxidant properties of activators of mitochondrial ATP-dependent potassium channels with the Amplex Red fluorescent indicator. APPL BIOCHEM MICRO+ 2013. [DOI: 10.1134/s0003683813040108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|