1
|
Fitts RH, Wang X, Kwok WM, Camara AKS. Cardiomyocyte Adaptation to Exercise: K+ Channels, Contractility and Ischemic Injury. Int J Sports Med 2024; 45:791-803. [PMID: 38648799 DOI: 10.1055/a-2296-7604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality, and exercise-training (TRN) is known to reduce risk factors and protect the heart from ischemia and reperfusion injury. Though the cardioprotective effects of exercise are well-documented, underlying mechanisms are not well understood. This review highlights recent findings and focuses on cardiac factors with emphasis on K+ channel control of the action potential duration (APD), β-adrenergic and adenosine regulation of cardiomyocyte function, and mitochondrial Ca2+ regulation. TRN-induced prolongation and shortening of the APD at low and high activation rates, respectively, is discussed in the context of a reduced response of the sarcolemma delayed rectifier potassium channel (IK) and increased content and activation of the sarcolemma KATP channel. A proposed mechanism underlying the latter is presented, including the phosphatidylinositol-3kinase/protein kinase B pathway. TRN induced increases in cardiomyocyte contractility and the response to adrenergic agonists are discussed. The TRN-induced protection from reperfusion injury is highlighted by the increased content and activation of the sarcolemma KATP channel and the increased phosphorylated glycogen synthase kinase-3β, which aid in preventing mitochondrial Ca2+ overload and mitochondria-triggered apoptosis. Finally, a brief section is presented on the increased incidences of atrial fibrillation associated with age and in life-long exercisers.
Collapse
Affiliation(s)
- Robert H Fitts
- Biological Sciences, Marquette University, Milwaukee, United States
| | - Xinrui Wang
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
| | - Wai-Meng Kwok
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
| | - Amadou K S Camara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
- Physiology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
2
|
Wexler AC, Dooge H, El-Meanawy S, Santos E, Hacker T, Tewari A, Alvarado FJ, Ramratnam M. Cardiac overexpression of a mitochondrial SUR2A splice variant impairs cardiac function and worsens myocardial ischemia reperfusion injury in female mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:10.1016/j.jmccpl.2024.100088. [PMID: 39507427 PMCID: PMC11539194 DOI: 10.1016/j.jmccpl.2024.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
The small splice variant of the sulfonylurea receptor protein isoform 2 A (SUR2A-55) targets mitochondria and enhances mitoKATP activity. In male mice the overexpression of this protein promotes cardioprotection, reducing myocardial injury after an ischemic insult. However, it is unclear what impact SUR2A-55 overexpression has on the female myocardium. To investigate the impact of SU2R2A-55 on the female heart, mice with cardiac specific transgenic overexpression of SUR2A-55 (TGSUR2A-55) were examined by resting echocardiography and histopathology. In addition, hearts were subjected to ischemia reperfusion (IR) injury. Female TGSUR2A-55 mice had resting LV dysfunction and worse hemodynamic recovery with increased infarct size after IR injury. RNA-seq analysis found 227 differential expressed genes between WT and TGSUR2A-55 female mouse hearts that were enriched in pathways of cellular metabolism. This was in direct contrast to male mice that had only four differentially expressed genes. Female TGSUR2A-55 mice compared to female WT mice had reduced cardiomyocyte mitochondrial membrane potential without a change in electron transport chain protein expression. In addition, isolated mitochondria from female TGSUR2A-55 hearts displayed reduced sensitivity to ATP and diazoxide suggestive of increased mitoKATP activity. In conclusion, our data suggests that female TGSUR2A-55 mice are unable to tolerate a more active mitoKATP channel leading to LV dysfunction and worse response to IR injury. This is in direct contrast to our prior report showing cardioprotection in male mice overexpressing SUR2A-55 in heart. Future research directed at examining the expression and activity of mitoKATP subunits according to sex may elucidate different treatments for male and female patients.
Collapse
Affiliation(s)
- Allison C. Wexler
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Holly Dooge
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Sarah El-Meanawy
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Elizabeth Santos
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Timothy Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Aditya Tewari
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Francisco J. Alvarado
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mohun Ramratnam
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| |
Collapse
|
3
|
Li K, Li Y, Chen Y, Chen T, Yang Y, Li P. Ion Channels Remodeling in the Regulation of Vascular Hyporesponsiveness During Shock. Microcirculation 2024; 31:e12874. [PMID: 39011763 DOI: 10.1111/micc.12874] [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: 07/19/2023] [Revised: 04/07/2024] [Accepted: 06/16/2024] [Indexed: 07/17/2024]
Abstract
Shock is characterized with vascular hyporesponsiveness to vasoconstrictors, thereby to cause refractory hypotension, insufficient tissue perfusion, and multiple organ dysfunction. The vascular hyporeactivity persisted even though norepinephrine and fluid resuscitation were administrated, it is of critical importance to find new potential target. Ion channels are crucial in the regulation of cell membrane potential and affect vasoconstriction and vasodilation. It has been demonstrated that many types of ion channels including K+ channels, Ca2+ permeable channels, and Na+ channels exist in vascular smooth muscle cells and endothelial cells, contributing to the regulation of vascular homeostasis and vasomotor function. An increasing number of studies suggested that the structural and functional alterations of ion channels located in arteries contribute to vascular hyporesponsiveness during shock, but the underlying mechanisms remained to be fully clarified. Therefore, the expression and functional changes in ion channels in arteries associated with shock are reviewed, to pave the way for further exploring the potential of ion channel-targeted compounds in treating refractory hypotension in shock.
Collapse
Affiliation(s)
- Keqing Li
- The Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuan Li
- The Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yinghong Chen
- The Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Tangting Chen
- The Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yan Yang
- The Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Pengyun Li
- The Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Domínguez Romero Y, Montoya Ortiz G, Novoa Herrán S, Osorio Mendez J, Gomez Grosso LA. miRNA Expression Profiles in Isolated Ventricular Cardiomyocytes: Insights into Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2024; 25:5272. [PMID: 38791311 PMCID: PMC11121573 DOI: 10.3390/ijms25105272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Doxorubicin (DOX), widely used as a chemotherapeutic agent for various cancers, is limited in its clinical utility by its cardiotoxic effects. Despite its widespread use, the precise mechanisms underlying DOX-induced cardiotoxicity at the cellular and molecular levels remain unclear, hindering the development of preventive and early detection strategies. To characterize the cytotoxic effects of DOX on isolated ventricular cardiomyocytes, focusing on the expression of specific microRNAs (miRNAs) and their molecular targets associated with endogenous cardioprotective mechanisms such as the ATP-sensitive potassium channel (KATP), Sirtuin 1 (SIRT1), FOXO1, and GSK3β. We isolated Guinea pig ventricular cardiomyocytes by retrograde perfusion and enzymatic dissociation. We assessed cell morphology, Reactive Oxygen Species (ROS) levels, intracellular calcium, and mitochondrial membrane potential using light microscopy and specific probes. We determined the miRNA expression profile using small RNAseq and validated it using stem-loop qRT-PCR. We quantified mRNA levels of some predicted and validated molecular targets using qRT-PCR and analyzed protein expression using Western blot. Exposure to 10 µM DOX resulted in cardiomyocyte shortening, increased ROS and intracellular calcium levels, mitochondrial membrane potential depolarization, and changes in specific miRNA expression. Additionally, we observed the differential expression of KATP subunits (ABCC9, KCNJ8, and KCNJ11), FOXO1, SIRT1, and GSK3β molecules associated with endogenous cardioprotective mechanisms. Supported by miRNA gene regulatory networks and functional enrichment analysis, these findings suggest that DOX-induced cardiotoxicity disrupts biological processes associated with cardioprotective mechanisms. Further research must clarify their specific molecular changes in DOX-induced cardiac dysfunction and investigate their diagnostic biomarkers and therapeutic potential.
Collapse
Affiliation(s)
- Yohana Domínguez Romero
- Doctorate in Biotechnology Program, Faculty of Sciences, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia; (G.M.O.); (S.N.H.); (J.O.M.)
| | - Gladis Montoya Ortiz
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia; (G.M.O.); (S.N.H.); (J.O.M.)
| | - Susana Novoa Herrán
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia; (G.M.O.); (S.N.H.); (J.O.M.)
| | - Jhon Osorio Mendez
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia; (G.M.O.); (S.N.H.); (J.O.M.)
- Master in Biochemistry Program, Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Luis A. Gomez Grosso
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia; (G.M.O.); (S.N.H.); (J.O.M.)
- Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| |
Collapse
|
5
|
Elangeeb ME, Elfaki I, Eleragi AMS, Ahmed EM, Mir R, Alzahrani SM, Bedaiwi RI, Alharbi ZM, Mir MM, Ajmal MR, Tayeb FJ, Barnawi J. Molecular Dynamics Simulation of Kir6.2 Variants Reveals Potential Association with Diabetes Mellitus. Molecules 2024; 29:1904. [PMID: 38675722 PMCID: PMC11054064 DOI: 10.3390/molecules29081904] [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: 03/07/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetes mellitus (DM) represents a problem for the healthcare system worldwide. DM has very serious complications such as blindness, kidney failure, and cardiovascular disease. In addition to the very bad socioeconomic impacts, it influences patients and their families and communities. The global costs of DM and its complications are huge and expected to rise by the year 2030. DM is caused by genetic and environmental risk factors. Genetic testing will aid in early diagnosis and identification of susceptible individuals or populations using ATP-sensitive potassium (KATP) channels present in different tissues such as the pancreas, myocardium, myocytes, and nervous tissues. The channels respond to different concentrations of blood sugar, stimulation by hormones, or ischemic conditions. In pancreatic cells, they regulate the secretion of insulin and glucagon. Mutations in the KCNJ11 gene that encodes the Kir6.2 protein (a major constituent of KATP channels) were reported to be associated with Type 2 DM, neonatal diabetes mellitus (NDM), and maturity-onset diabetes of the young (MODY). Kir6.2 harbors binding sites for ATP and phosphatidylinositol 4,5-diphosphate (PIP2). The ATP inhibits the KATP channel, while the (PIP2) activates it. A Kir6.2 mutation at tyrosine330 (Y330) was demonstrated to reduce ATP inhibition and predisposes to NDM. In this study, we examined the effect of mutations on the Kir6.2 structure using bioinformatics tools and molecular dynamic simulations (SIFT, PolyPhen, SNAP2, PANTHER, PhD&SNP, SNP&Go, I-Mutant, MuPro, MutPred, ConSurf, HOPE, and GROMACS). Our results indicated that M199R, R201H, R206H, and Y330H mutations influence Kir6.2 structure and function and therefore may cause DM. We conclude that MD simulations are useful techniques to predict the effects of mutations on protein structure. In addition, the M199R, R201H, R206H, and Y330H variant in the Kir6.2 protein may be associated with DM. These results require further verification in protein-protein interactions, Kir6.2 function, and case-control studies.
Collapse
Affiliation(s)
- Mohamed E. Elangeeb
- Department of Basic Medical Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.M.A.); (M.R.A.)
| | - Ali M. S. Eleragi
- Department of Microbiology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Elsadig Mohamed Ahmed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia;
- Department of Clinical Chemistry, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti 27711, Sudan
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (R.I.B.); (Z.M.A.); (F.J.T.); (J.B.)
| | - Salem M. Alzahrani
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.M.A.); (M.R.A.)
| | - Ruqaiah I. Bedaiwi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (R.I.B.); (Z.M.A.); (F.J.T.); (J.B.)
| | - Zeyad M. Alharbi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (R.I.B.); (Z.M.A.); (F.J.T.); (J.B.)
| | - Mohammad Muzaffar Mir
- Department of Clinical Biochemistry, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Mohammad Rehan Ajmal
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; (S.M.A.); (M.R.A.)
| | - Faris Jamal Tayeb
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (R.I.B.); (Z.M.A.); (F.J.T.); (J.B.)
| | - Jameel Barnawi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.M.); (R.I.B.); (Z.M.A.); (F.J.T.); (J.B.)
| |
Collapse
|
6
|
Chinawa JM, Chinawa AT, Chukwu BF, Onyia JT. Assessment of descending aortic blood flow velocities with continuous wave Doppler echocardiography among healthy Children in South East Nigeria. Malawi Med J 2024; 36:1-6. [PMID: 39086365 PMCID: PMC11287811 DOI: 10.4314/mmj.v36i.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Abstract
Background The descending aorta velocity is important predictor of aortic disease in children and can be very helpful in some clinical and surgical decision making. Aim The purpose of this study is to assess the normative values of descending aorta velocity among children from South-East Nigeria. It also aimed to assess the correlation between age, body surface area and mean velocity across the descending aorta. Methods This is a cross-sectional study where the descending aorta velocity of one hundred and eleven children were enrolled consecutively using digitized two-dimensional and Doppler echocardiography. Results A total of 111 children had echocardiography to study their cardiac structures and compute their mean scores of their descending aorta velocity. The mean velocity across the descending aorta was 1.3±0.2m/s with maximum and minimum velocities of 2.06 and 0.84cm respectively. The mean descending aorta velocity in males (1.37±0.24 m/s) was significantly higher than that in females (1.24±0.18); (Student T test 3.09, p = 0.03). There was no correlation between age and mean velocity across the descending aorta (Pearson correlation coefficient; -0.03, p = 0.7) nor between body surface area and descending aorta velocity (correlation coefficient 0.01, p= 0.8). Conclusions The presented normalized values of the descending aorta velocity using a digitized two-dimensional and Doppler echocardiography among healthy children will serve as a reference values for further studies and can be applied for clinical and surgical use in children with various cardiac anomalies.
Collapse
Affiliation(s)
- Josephat M Chinawa
- Department of Paediatrics, College of Medicine, University of Nigeria Ituku/Ozalla and University of Nigeria Teaching Hospital Ituku/Ozalla, Enugu, Enugu State, Nigeria
| | - Awoere T Chinawa
- Department of Community Medicine, ESUCOM, Parklane Enugu, Enugu State, Nigeria
| | | | - Jude T Onyia
- Department of Community Medicine, ESUCOM, Parklane Enugu, Enugu State, Nigeria
| |
Collapse
|
7
|
Longden TA, Lederer WJ. Electro-metabolic signaling. J Gen Physiol 2024; 156:e202313451. [PMID: 38197953 PMCID: PMC10783436 DOI: 10.1085/jgp.202313451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Precise matching of energy substrate delivery to local metabolic needs is essential for the health and function of all tissues. Here, we outline a mechanistic framework for understanding this critical process, which we refer to as electro-metabolic signaling (EMS). All tissues exhibit changes in metabolism over varying spatiotemporal scales and have widely varying energetic needs and reserves. We propose that across tissues, common signatures of elevated metabolism or increases in energy substrate usage that exceed key local thresholds rapidly engage mechanisms that generate hyperpolarizing electrical signals in capillaries that then relax contractile elements throughout the vasculature to quickly adjust blood flow to meet changing needs. The attendant increase in energy substrate delivery serves to meet local metabolic requirements and thus avoids a mismatch in supply and demand and prevents metabolic stress. We discuss in detail key examples of EMS that our laboratories have discovered in the brain and the heart, and we outline potential further EMS mechanisms operating in tissues such as skeletal muscle, pancreas, and kidney. We suggest that the energy imbalance evoked by EMS uncoupling may be central to cellular dysfunction from which the hallmarks of aging and metabolic diseases emerge and may lead to generalized organ failure states-such as diverse flavors of heart failure and dementia. Understanding and manipulating EMS may be key to preventing or reversing these dysfunctions.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W. Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Balistrieri A, Makino A, Yuan JXJ. Pathophysiology and pathogenic mechanisms of pulmonary hypertension: role of membrane receptors, ion channels, and Ca 2+ signaling. Physiol Rev 2023; 103:1827-1897. [PMID: 36422993 PMCID: PMC10110735 DOI: 10.1152/physrev.00030.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The pulmonary circulation is a low-resistance, low-pressure, and high-compliance system that allows the lungs to receive the entire cardiac output. Pulmonary arterial pressure is a function of cardiac output and pulmonary vascular resistance, and pulmonary vascular resistance is inversely proportional to the fourth power of the intraluminal radius of the pulmonary artery. Therefore, a very small decrease of the pulmonary vascular lumen diameter results in a significant increase in pulmonary vascular resistance and pulmonary arterial pressure. Pulmonary arterial hypertension is a fatal and progressive disease with poor prognosis. Regardless of the initial pathogenic triggers, sustained pulmonary vasoconstriction, concentric vascular remodeling, occlusive intimal lesions, in situ thrombosis, and vascular wall stiffening are the major and direct causes for elevated pulmonary vascular resistance in patients with pulmonary arterial hypertension and other forms of precapillary pulmonary hypertension. In this review, we aim to discuss the basic principles and physiological mechanisms involved in the regulation of lung vascular hemodynamics and pulmonary vascular function, the changes in the pulmonary vasculature that contribute to the increased vascular resistance and arterial pressure, and the pathogenic mechanisms involved in the development and progression of pulmonary hypertension. We focus on reviewing the pathogenic roles of membrane receptors, ion channels, and intracellular Ca2+ signaling in pulmonary vascular smooth muscle cells in the development and progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- Harvard University, Cambridge, Massachusetts
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
9
|
McClenaghan C, Mukadam MA, Roeglin J, Tryon RC, Grabner M, Dayal A, Meyer GA, Nichols CG. Skeletal muscle delimited myopathy and verapamil toxicity in SUR2 mutant mouse models of AIMS. EMBO Mol Med 2023; 15:e16883. [PMID: 37154692 PMCID: PMC10245035 DOI: 10.15252/emmm.202216883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
ABCC9-related intellectual disability and myopathy syndrome (AIMS) arises from loss-of-function (LoF) mutations in the ABCC9 gene, which encodes the SUR2 subunit of ATP-sensitive potassium (KATP ) channels. KATP channels are found throughout the cardiovascular system and skeletal muscle and couple cellular metabolism to excitability. AIMS individuals show fatigability, muscle spasms, and cardiac dysfunction. We found reduced exercise performance in mouse models of AIMS harboring premature stop codons in ABCC9. Given the roles of KATP channels in all muscles, we sought to determine how myopathy arises using tissue-selective suppression of KATP and found that LoF in skeletal muscle, specifically, underlies myopathy. In isolated muscle, SUR2 LoF results in abnormal generation of unstimulated forces, potentially explaining painful spasms in AIMS. We sought to determine whether excessive Ca2+ influx through CaV 1.1 channels was responsible for myopathology but found that the Ca2+ channel blocker verapamil unexpectedly resulted in premature death of AIMS mice and that rendering CaV 1.1 channels nonpermeable by mutation failed to reverse pathology; results which caution against the use of calcium channel blockers in AIMS.
Collapse
Affiliation(s)
- Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| | - Maya A Mukadam
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Jacob Roeglin
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Robert C Tryon
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Manfred Grabner
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Anamika Dayal
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Gretchen A Meyer
- Program in Physical Therapy, Departments of Orthopaedic Surgery, Neurology and Biomedical EngineeringWashington University School of MedicineSt. LouisMOUSA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
10
|
Strutynskyi RB, Strutynska NA, Piven OO, Mys LA, Goshovska YV, Fedichkina RA, Okhai IY, Strutynskyi VR, Dosenko VE, Dobrzyn P, Sagach VF. Upregulation of ATP-Sensitive Potassium Channels as the Potential Mechanism of Cardioprotection and Vasorelaxation Under the Action of Pyridoxal-5-Phosphate in Old Rats. J Cardiovasc Pharmacol Ther 2023; 28:10742484231213175. [PMID: 37946524 DOI: 10.1177/10742484231213175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Background: The aging process is accompanied by the weakening of the protective systems of the organism, in particular by the decrease in the expression of ATP-sensitive potassium (KATP) channels and in the synthesis of H2S. The aim of our work was to investigate the role of KATP channels in the cardioprotection induced by pyridoxal-5-phosphate (PLP) in aging. Methods: Experiments were performed on adult and old (aged 24 months) male Wistar rats, which were divided into 3 groups: adults, old, and old PLP-treated rats. PLP was administered orally once a day for 14 days at a dose of 0.7 mg/kg. The levels of mRNA expression of subunits KATP channels were determined by reverse transcription and real-time polymerase chain reaction analysis. Protein expression levels were determined by the Western blot. Cardiac tissue morphology was determined using transverse 6 μm deparaffinized sections stained with picrosirius red staining. Vasorelaxation responses of isolated aortic rings and the function of Langendorff-perfused isolated hearts during ischemia-reperfusion, H2S levels, and markers of oxidative stress were also studied. Results: Administration of PLP to old rats reduces cardiac fibrosis and improves cardiac function during ischemia-reperfusion and vasorelaxation responses to KATP channels opening. At the same time, there was a significant increase in mRNA and protein expression of SUR2 and Kir6.1 subunits of KATP channels, H2S production, and reduced markers of oxidative stress. The specific KATP channel inhibitor-glibenclamide prevented the enhancement of vasodilator responses and anti-ischemic protection in PLP-treated animals. Conclusions: We suggest that this potential therapeutic effect of PLP in old animals may be a result of increased expression of KATP channels and H2S production.
Collapse
Affiliation(s)
- Ruslan B Strutynskyi
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Nataliіa A Strutynska
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Oksana O Piven
- The Laboratory of Molecular Medical Biochemistry of Nencki, Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Lidiia A Mys
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Yulia V Goshovska
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Raisa A Fedichkina
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Iryna Y Okhai
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Vladyslav R Strutynskyi
- Department of Immunophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Victor E Dosenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Pawel Dobrzyn
- The Laboratory of Molecular Medical Biochemistry of Nencki, Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Vadim F Sagach
- Department of Blood Circulation, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
11
|
Younis H, Ha SE, Jorgensen BG, Verma A, Ro S. Maturity-Onset Diabetes of the Young: Mutations, Physiological Consequences, and Treatment Options. J Pers Med 2022; 12:jpm12111762. [PMID: 36573710 PMCID: PMC9697644 DOI: 10.3390/jpm12111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 02/01/2023] Open
Abstract
Maturity-Onset Diabetes of the Young (MODY) is a rare form of diabetes which affects between 1% and 5% of diagnosed diabetes cases. Clinical characterizations of MODY include onset of diabetes at an early age (before the age of 30), autosomal dominant inheritance pattern, impaired glucose-induced secretion of insulin, and hyperglycemia. Presently, 14 MODY subtypes have been identified. Within these subtypes are several mutations which contribute to the different MODY phenotypes. Despite the identification of these 14 subtypes, MODY is often misdiagnosed as type 1 or type 2 diabetes mellitus due to an overlap in clinical features, high cost and limited availability of genetic testing, and unfamiliarity with MODY outside of the medical profession. The primary aim of this review is to investigate the genetic characterization of the MODY subtypes. Additionally, this review will elucidate the link between the genetics, function, and clinical manifestations of MODY in each of the 14 subtypes. In providing this knowledge, we hope to assist in the accurate diagnosis of MODY patients and, subsequently, in ensuring they receive appropriate treatment.
Collapse
Affiliation(s)
- Hazar Younis
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Arushi Verma
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- RosVivo Therapeutics, Applied Research Facility, Reno, NV 89557, USA
- Correspondence:
| |
Collapse
|
12
|
McClenaghan C, Nichols CG. Kir6.1 and SUR2B in Cantú syndrome. Am J Physiol Cell Physiol 2022; 323:C920-C935. [PMID: 35876283 PMCID: PMC9467476 DOI: 10.1152/ajpcell.00154.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 12/25/2022]
Abstract
Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?
Collapse
Affiliation(s)
- Conor McClenaghan
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| |
Collapse
|
13
|
Wang Z, Bian W, Yan Y, Zhang DM. Functional Regulation of KATP Channels and Mutant Insight Into Clinical Therapeutic Strategies in Cardiovascular Diseases. Front Pharmacol 2022; 13:868401. [PMID: 35837280 PMCID: PMC9274113 DOI: 10.3389/fphar.2022.868401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
ATP-sensitive potassium channels (KATP channels) play pivotal roles in excitable cells and link cellular metabolism with membrane excitability. The action potential converts electricity into dynamics by ion channel-mediated ion exchange to generate systole, involved in every heartbeat. Activation of the KATP channel repolarizes the membrane potential and decreases early afterdepolarization (EAD)-mediated arrhythmias. KATP channels in cardiomyocytes have less function under physiological conditions but they open during severe and prolonged anoxia due to a reduced ATP/ADP ratio, lessening cellular excitability and thus preventing action potential generation and cell contraction. Small active molecules activate and enhance the opening of the KATP channel, which induces the repolarization of the membrane and decreases the occurrence of malignant arrhythmia. Accumulated evidence indicates that mutation of KATP channels deteriorates the regulatory roles in mutation-related diseases. However, patients with mutations in KATP channels still have no efficient treatment. Hence, in this study, we describe the role of KATP channels and subunits in angiocardiopathy, summarize the mutations of the KATP channels and the functional regulation of small active molecules in KATP channels, elucidate the potential mechanisms of mutant KATP channels and provide insight into clinical therapeutic strategies.
Collapse
Affiliation(s)
- Zhicheng Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Weikang Bian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yufeng Yan
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Dai-Min Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Dai-Min Zhang,
| |
Collapse
|
14
|
Yang HQ, Echeverry FA, ElSheikh A, Gando I, Anez Arredondo S, Samper N, Cardozo T, Delmar M, Shyng SL, Coetzee WA. Subcellular trafficking and endocytic recycling of K ATP channels. Am J Physiol Cell Physiol 2022; 322:C1230-C1247. [PMID: 35508187 PMCID: PMC9169827 DOI: 10.1152/ajpcell.00099.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 11/22/2022]
Abstract
Sarcolemmal/plasmalemmal ATP-sensitive K+ (KATP) channels have key roles in many cell types and tissues. Hundreds of studies have described how the KATP channel activity and ATP sensitivity can be regulated by changes in the cellular metabolic state, by receptor signaling pathways and by pharmacological interventions. These alterations in channel activity directly translate to alterations in cell or tissue function, that can range from modulating secretory responses, such as insulin release from pancreatic β-cells or neurotransmitters from neurons, to modulating contractile behavior of smooth muscle or cardiac cells to elicit alterations in blood flow or cardiac contractility. It is increasingly becoming apparent, however, that KATP channels are regulated beyond changes in their activity. Recent studies have highlighted that KATP channel surface expression is a tightly regulated process with similar implications in health and disease. The surface expression of KATP channels is finely balanced by several trafficking steps including synthesis, assembly, anterograde trafficking, membrane anchoring, endocytosis, endocytic recycling, and degradation. This review aims to summarize the physiological and pathophysiological implications of KATP channel trafficking and mechanisms that regulate KATP channel trafficking. A better understanding of this topic has potential to identify new approaches to develop therapeutically useful drugs to treat KATP channel-related diseases.
Collapse
Affiliation(s)
- Hua-Qian Yang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China
| | | | - Assmaa ElSheikh
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
- Department of Medical Biochemistry, Tanta University, Tanta, Egypt
| | - Ivan Gando
- Department of Pathology, NYU School of Medicine, New York, New York
| | | | - Natalie Samper
- Department of Pathology, NYU School of Medicine, New York, New York
| | - Timothy Cardozo
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - Mario Delmar
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
- Department of Medicine, NYU School of Medicine, New York, New York
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| | - William A Coetzee
- Department of Pathology, NYU School of Medicine, New York, New York
- Department of Neuroscience & Physiology, NYU School of Medicine, New York, New York
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
15
|
Gorshkova OP. Age-Related Changes in the Functional Activity of ATP-Sensitive Potassium Channels in Rat Pial Arteries. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020041] [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]
|
16
|
Ando K, Tong L, Peng D, Vázquez-Liébanas E, Chiyoda H, He L, Liu J, Kawakami K, Mochizuki N, Fukuhara S, Grutzendler J, Betsholtz C. KCNJ8/ABCC9-containing K-ATP channel modulates brain vascular smooth muscle development and neurovascular coupling. Dev Cell 2022; 57:1383-1399.e7. [PMID: 35588738 DOI: 10.1016/j.devcel.2022.04.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/22/2021] [Accepted: 04/26/2022] [Indexed: 12/30/2022]
Abstract
Loss- or gain-of-function mutations in ATP-sensitive potassium channel (K-ATP)-encoding genes, KCNJ8 and ABCC9, cause human central nervous system disorders with unknown pathogenesis. Here, using mice, zebrafish, and cell culture models, we investigated cellular and molecular causes of brain dysfunctions derived from altered K-ATP channel function. We show that genetic/chemical inhibition or activation of KCNJ8/ABCC9-containing K-ATP channel function leads to brain-selective suppression or promotion of arterial/arteriolar vascular smooth muscle cell (VSMC) differentiation, respectively. We further show that brain VSMCs develop from KCNJ8/ABCC9-containing K-ATP channel-expressing mural cell progenitor and that K-ATP channel cell autonomously regulates VSMC differentiation through modulation of intracellular Ca2+ oscillation via voltage-dependent calcium channels. Consistent with defective VSMC development, Kcnj8 knockout mice showed deficiency in vasoconstrictive capacity and neuronal-evoked vasodilation leading to local hyperemia. Our results demonstrate a role for KCNJ8/ABCC9-containing K-ATP channels in the differentiation of brain VSMC, which in turn is necessary for fine-tuning of cerebral blood flow.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan.
| | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Hirohisa Chiyoda
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Jianping Liu
- Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden.
| |
Collapse
|
17
|
Transcriptome-Guided Identification of Drugs for Repurposing to Treat Age-Related Hearing Loss. Biomolecules 2022; 12:biom12040498. [PMID: 35454087 PMCID: PMC9028743 DOI: 10.3390/biom12040498] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Age-related hearing loss (ARHL) or presbycusis is a prevalent condition associated with social isolation, cognitive impairment, and dementia. Age-related changes in the cochlea, the auditory portion of the inner ear, are the primary cause of ARHL. Unfortunately, there are currently no pharmaceutical approaches to treat ARHL. To examine the biological processes underlying age-related changes in the cochlea and identify candidate drugs for rapid repurposing to treat ARHL, we utilized bulk RNA sequencing to obtain transcriptomes from the functional substructures of the cochlea—the sensorineural structures, including the organ of Corti and spiral ganglion neurons (OC/SGN) and the stria vascularis and spiral ligament (SV/SL)—in young (6-week-old) and old (2-year-old) C57BL/6 mice. Transcriptomic analyses revealed both overlapping and unique patterns of gene expression and gene enrichment between substructures and with ageing. Based on these age-related transcriptional changes, we queried the protein products of genes differentially expressed with ageing in DrugBank and identified 27 FDA/EMA-approved drugs that are suitable to be repurposed to treat ARHL. These drugs target the protein products of genes that are differentially expressed with ageing uniquely in either the OC/SGN or SV/SL and that interrelate diverse biological processes. Further transcriptomic analyses revealed that most genes differentially expressed with ageing in both substructures encode protein products that are promising drug target candidates but are, nevertheless, not yet linked to approved drugs. Thus, with this study, we apply a novel approach to characterize the druggable genetic landscape for ARHL and propose a list of drugs to test in pre-clinical studies as potential treatment options for ARHL.
Collapse
|
18
|
Alquisiras-Burgos I, Franco-Pérez J, Rubio-Osornio M, Aguilera P. The short form of the SUR1 and its functional implications in the damaged brain. Neural Regen Res 2022; 17:488-496. [PMID: 34380876 PMCID: PMC8504400 DOI: 10.4103/1673-5374.320967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Sulfonylurea receptor (SUR) belongs to the adenosine 5′-triphosphate (ATP)-binding cassette (ABC) transporter family; however, SUR is associated with ion channels and acts as a regulatory subunit determining the opening or closing of the pore. Abcc8 and Abcc9 genes code for the proteins SUR1 and SUR2, respectively. The SUR1 transcript encodes a protein of 1582 amino acids with a mass around 140–177 kDa expressed in the pancreas, brain, heart, and other tissues. It is well known that SUR1 assembles with Kir6.2 and TRPM4 to establish KATP channels and non-selective cation channels, respectively. Abbc8 and 9 are alternatively spliced, and the resulting transcripts encode different isoforms of SUR1 and SUR2, which have been detected by different experimental strategies. Interestingly, the use of binding assays to sulfonylureas and Western blotting has allowed the detection of shorter forms of SUR (~65 kDa). Identity of the SUR1 variants has not been clarified, and some authors have suggested that the shorter forms are unspecific. However, immunoprecipitation assays have shown that SUR2 short forms are part of a functional channel even coexisting with the typical forms of the receptor in the heart. This evidence confirms that the structure of the short forms of the SURs is fully functional and does not lose the ability to interact with the channels. Since structural changes in short forms of SUR modify its affinity to ATP, regulation of its expression might represent an advantage in pathologies where ATP concentrations decrease and a therapeutic target to induce neuroprotection. Remarkably, the expression of SUR1 variants might be induced by conditions associated to the decrease of energetic substrates in the brain (e.g. during stroke and epilepsy). In this review, we want to contribute to the knowledge of SUR1 complexity by analyzing evidence that shows the existence of short SUR1 variants and its possible implications in brain function.
Collapse
Affiliation(s)
- Iván Alquisiras-Burgos
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| | - Javier Franco-Pérez
- Laboratorio de Formación Reticular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| | - Moisés Rubio-Osornio
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| | - Penélope Aguilera
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", CDMX, Mexico
| |
Collapse
|
19
|
Vascular K ATP channel structural dynamics reveal regulatory mechanism by Mg-nucleotides. Proc Natl Acad Sci U S A 2021; 118:2109441118. [PMID: 34711681 PMCID: PMC8694068 DOI: 10.1073/pnas.2109441118] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 11/24/2022] Open
Abstract
Vascular KATP channels formed by the potassium channel Kir6.1 and its regulatory protein SUR2B maintain blood pressure in the physiological range. Overactivity of the channel due to genetic mutations in either Kir6.1 or SUR2B causes severe cardiovascular pathologies known as Cantú syndrome. The cryogenic electron microscopy structures of the vascular KATP channel reported here show multiple, dynamically related conformations of the regulatory subunit SUR2B. Molecular dynamics simulations reveal the negatively charged ED-domain in SUR2B, a stretch of 15 glutamate (E) and aspartate (D) residues not previously resolved, play a key MgADP-dependent role in mediating interactions at the interface between the SUR2B and Kir6.1 subunits. Our findings provide a mechanistic understanding of how channel activity is regulated by intracellular MgADP. Vascular tone is dependent on smooth muscle KATP channels comprising pore-forming Kir6.1 and regulatory SUR2B subunits, in which mutations cause Cantú syndrome. Unique among KATP isoforms, they lack spontaneous activity and require Mg-nucleotides for activation. Structural mechanisms underlying these properties are unknown. Here, we determined cryogenic electron microscopy structures of vascular KATP channels bound to inhibitory ATP and glibenclamide, which differ informatively from similarly determined pancreatic KATP channel isoform (Kir6.2/SUR1). Unlike SUR1, SUR2B subunits adopt distinct rotational “propeller” and “quatrefoil” geometries surrounding their Kir6.1 core. The glutamate/aspartate-rich linker connecting the two halves of the SUR-ABC core is observed in a quatrefoil-like conformation. Molecular dynamics simulations reveal MgADP-dependent dynamic tripartite interactions between this linker, SUR2B, and Kir6.1. The structures captured implicate a progression of intermediate states between MgADP-free inactivated, and MgADP-bound activated conformations wherein the glutamate/aspartate-rich linker participates as mobile autoinhibitory domain, suggesting a conformational pathway toward KATP channel activation.
Collapse
|
20
|
Gorshkova OP, Sokolova IB. Ischemia Changes the Contribution of KATP-Channels to Basal Tone and Dilation of Rat Pial Arteries. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021050136] [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]
|
21
|
The expression of ATP-sensitive potassium channels in human umbilical arteries with severe pre-eclampsia. Sci Rep 2021; 11:7955. [PMID: 33846486 PMCID: PMC8041753 DOI: 10.1038/s41598-021-87146-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/22/2021] [Indexed: 11/29/2022] Open
Abstract
The aim of this study is to establish the expression of ATP-sensitive potassium channels(KATP) in human umbilical arteries with severe pre-eclampsia. Real-time quantitative PCR and western blotting were used to detect the mRNA and protein expression levels of KATP channel subunits Kir6.1 and SUR2B in human umbilical arteries from normal pregnant and those with severe pre-eclampsia, early onset severe pre-eclampsia and late onset severe pre-eclampsia. The mRNA and protein levels of SUR2B in the severe pre-eclampsia group were lower than those in the normal group (P < 0.001), and the expression of Kir6.1 was not statistically significant between the two groups (P > 0.05). The mRNA and protein levels of SUR2B in early onset severe pre-eclampsia group were lower than those in late onset severe pre-eclampsia group (P < 0.001). There was no significant difference in expression of Kir6.1 between the two groups (P > 0.05). The mRNA and protein expression levels of SUR2B in pregnant women with severe pre-eclampsia were lower than those in normal pregnant women, suggesting that the expression of the SUR2B of the KATP channel may be related to the occurrence and development of severe pre-eclampsia. Compared with late onset severe pre-eclampsia, the mRNA and protein expression levels of SUR2B were lower in the umbilical arteries of women with early onset severe pre-eclampsia, suggesting that the occurrence time of severe pre-eclampsia may be related to the extent reduced expression of the SUR2B of the KATP channel.
Collapse
|
22
|
Nii T, Eguchi R, Yamaguchi S, Otsuguro KI. Hydrogen sulfide induces Ca 2+ release from the endoplasmic reticulum and suppresses ATP-induced Ca 2+ signaling in rat spinal cord astrocytes. Eur J Pharmacol 2021; 891:173684. [PMID: 33129788 DOI: 10.1016/j.ejphar.2020.173684] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
Hydrogen sulfide (H2S) has a variety of physiological functions. H2S reportedly increases intracellular Ca2+ concentration ([Ca2+]i) in astrocytes. However, the precise mechanism and functional role of this increase are not known. Here, we examined the effects of H2S on [Ca2+]i in astrocytes from the rat spinal cord and whether H2S affects ATP-induced Ca2+ signaling, which is known to be involved in synaptic function. Na2S (150 μM), an H2S donor, produced a nontoxic increase in [Ca2+]i. The [Ca2+]i increase by Na2S was inhibited by Ca2+ depletion in the endoplasmic reticulum (ER) but not by removal of extracellular Ca2+, indicating that H2S releases Ca2+ from the ER. On the other hand, Na2S inhibited ATP-induced [Ca2+]i increase when Na2S clearly increased [Ca2+]i in the astrocytes, which was not suppressed by a reducing agent. In addition, Na2S had no effect on intracellular cyclic AMP (cAMP) level. These results indicate that oxidative post-translational modification of proteins and cAMP are not involved in the inhibitory effect of H2S on ATP-induced Ca2+ signaling. We conclude that H2S indirectly inhibits ATP-induced Ca2+ signaling by decreasing Ca2+ content in the ER in astrocytes. In this way, H2S may influence intercellular communication between astrocytes and neurons, thereby contributing to neuronal signaling in the nervous system.
Collapse
Affiliation(s)
- Takeshi Nii
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Ryota Eguchi
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Soichiro Yamaguchi
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Ken-Ichi Otsuguro
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan.
| |
Collapse
|
23
|
Clinical Importance of the Human Umbilical Artery Potassium Channels. Cells 2020; 9:cells9091956. [PMID: 32854241 PMCID: PMC7565333 DOI: 10.3390/cells9091956] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Potassium (K+) channels are usually predominant in the membranes of vascular smooth muscle cells (SMCs). These channels play an important role in regulating the membrane potential and vessel contractility-a role that depends on the vascular bed. Thus, the activity of K+ channels represents one of the main mechanisms regulating the vascular tone in physiological and pathophysiological conditions. Briefly, the activation of K+ channels in SMC leads to hyperpolarization and vasorelaxation, while its inhibition induces depolarization and consequent vascular contraction. Currently, there are four different types of K+ channels described in SMCs: voltage-dependent K+ (KV) channels, calcium-activated K+ (KCa) channels, inward rectifier K+ (Kir) channels, and 2-pore domain K+ (K2P) channels. Due to the fundamental role of K+ channels in excitable cells, these channels are promising therapeutic targets in clinical practice. Therefore, this review discusses the basic properties of the various types of K+ channels, including structure, cellular mechanisms that regulate their activity, and new advances in the development of activators and blockers of these channels. The vascular functions of these channels will be discussed with a focus on vascular SMCs of the human umbilical artery. Then, the clinical importance of K+ channels in the treatment and prevention of cardiovascular diseases during pregnancy, such as gestational hypertension and preeclampsia, will be explored.
Collapse
|
24
|
Zhang Q, Dou H, Rorsman P. 'Resistance is futile?' - paradoxical inhibitory effects of K ATP channel closure in glucagon-secreting α-cells. J Physiol 2020; 598:4765-4780. [PMID: 32716554 PMCID: PMC7689873 DOI: 10.1113/jp279775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
By secreting insulin and glucagon, the β- and α-cells of the pancreatic islets play a central role in the regulation of systemic metabolism. Both cells are equipped with ATP-regulated potassium (KATP ) channels that are regulated by the intracellular ATP/ADP ratio. In β-cells, KATP channels are active at low (non-insulin-releasing) glucose concentrations. An increase in glucose leads to KATP channel closure, membrane depolarization and electrical activity that culminates in elevation of [Ca2+ ]i and initiation of exocytosis of the insulin-containing secretory granules. The α-cells are also equipped with KATP channels but they are under strong tonic inhibition at low glucose, explaining why α-cells are electrically active under hypoglycaemic conditions and generate large Na+ - and Ca2+ -dependent action potentials. Closure of residual KATP channel activity leads to membrane depolarization and an increase in action potential firing but this stimulation of electrical activity is associated with inhibition rather than acceleration of glucagon secretion. This paradox arises because membrane depolarization reduces the amplitude of the action potentials by voltage-dependent inactivation of the Na+ channels involved in action potential generation. Exocytosis in α-cells is tightly linked to the opening of voltage-gated P/Q-type Ca2+ channels, the activation of which is steeply voltage-dependent. Accordingly, the inhibitory effect of the reduced action potential amplitude exceeds the stimulatory effect resulting from the increased action potential frequency. These observations highlight a previously unrecognised role of the action potential amplitude as a key regulator of pancreatic islet hormone secretion.
Collapse
Affiliation(s)
- Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Haiqiang Dou
- Metabolic Physiology Unit, Institute of Neuroscience and Physiology, University of Göteborg, PO Box 430, Göteborg, SE-405 30, Sweden
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK.,Metabolic Physiology Unit, Institute of Neuroscience and Physiology, University of Göteborg, PO Box 430, Göteborg, SE-405 30, Sweden
| |
Collapse
|
25
|
Akyuz E, Villa C. A novel role of cardiac inwardly rectifying potassium channels explaining autonomic cardiovascular dysfunctions in a cuprizone-induced mouse model of multiple sclerosis. Auton Neurosci 2020; 225:102647. [PMID: 32087534 DOI: 10.1016/j.autneu.2020.102647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), believed to have an autoimmune etiology. MS patients showed an increased cardiovascular (CV) risk probably related to an impairment in the autonomic control of CV functions, but the underlying molecular mechanisms are not completely elucidated. Inwardly-rectifying potassium (Kir) channels play a key role in cardiac excitability by contributing to the repolarization phase of action potential and were recently identified as target of the autoantibody response in MS patients. Therefore, we investigated the role of cardiac Kir channels in the CV dysfunctions occurring in MS. Cardiac functions were evaluated by electrocardiographic recordings (ECG) in cuprizone-fed C57BL/6 mice, a classic demyelination animal model. Gene expression profiling of cardiac Kir2.2, Kir4.1 and Kir6.2 channels was performed using real-time PCR in mice. Cuprizone-induced mouse model was confirmed by immunohistochemistry analysis showing demyelination in the corpus callosum. ECG recordings from mice showed a significant decreased duration of the P wave and RR interval as well as an increase of the heart rate in cuprizone-treated mice as compared with the controls. Significant increased relative expression levels of Kcnj11 and Kcnj12, encoding for Kir6.2 and Kir2.2 channels respectively, were observed in mouse heart tissue, whereas no differences were found in mRNA levels of Kir4.1 channel as compared with controls. For the first time, these findings provided valuable insights into the potential role of Kir channels in cardiac problems associated with MS.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey.
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| |
Collapse
|
26
|
Protective effect of Xin-Ji-Er-Kang on cardiovascular remodeling in high-salt induced hypertensive mice: Role ofoxidative stress and endothelial dysfunction. Biomed Pharmacother 2019; 115:108937. [DOI: 10.1016/j.biopha.2019.108937] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 11/18/2022] Open
|