101
|
Nikolaienko R, Bovo E, Rebbeck RT, Kahn D, Thomas DD, Cornea RL, Zima AV. The functional significance of redox-mediated intersubunit cross-linking in regulation of human type 2 ryanodine receptor. Redox Biol 2020; 37:101729. [PMID: 32980662 PMCID: PMC7522892 DOI: 10.1016/j.redox.2020.101729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/19/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
The type 2 ryanodine receptor (RyR2) plays a key role in the cardiac intracellular calcium (Ca2+) regulation. We have previously shown that oxidative stress activates RyR2 in rabbit cardiomyocytes by promoting the formation of disulfide bonds between neighboring RyR2 subunits. However, the functional significance of this redox modification for human RyR2 (hRyR2) remains largely unknown. Here, we studied the redox regulation of hRyR2 in HEK293 cells transiently expressing the ryr2 gene. Analysis of hRyR2 cross-linking and of the redox-GFP readout response to diamide oxidation revealed that hRyR2 cysteines involved in the intersubunit cross-linking are highly sensitive to oxidative stress. In parallel experiments, the effect of diamide on endoplasmic reticulum (ER) Ca2+ release was studied in cells co-transfected with hRyR2, ER Ca2+ pump (SERCA2a) and the ER-targeted Ca2+ sensor R-CEPIA1er. Expression of hRyR2 and SERCA2a produced “cardiac-like” Ca2+ waves due to spontaneous hRyR2 activation. Incubation with diamide caused a fast decline of the luminal ER Ca2+ (or ER Ca2+ load) followed by the cessation of Ca2+ waves. The maximal effect of diamide on ER Ca2+ load and Ca2+ waves positively correlates with the maximum level of hRyR2 cross-linking, indicating a functional significance of this redox modification. Furthermore, the level of hRyR2 cross-linking positively correlates with the degree of calmodulin (CaM) dissociation from the hRyR2 complex. In skeletal muscle RyR (RyR1), cysteine 3635 (C3635) is viewed as dominantly responsible for the redox regulation of the channel. Here, we showed that the corresponding cysteine 3602 (C3602) in hRyR2 does not participate in intersubunit cross-linking and plays a limited role in the hRyR2 regulation by CaM during oxidative stress. Collectively, these results suggest that redox-mediated intersubunit cross-linking is an important regulator of hRyR2 function under pathological conditions associated with oxidative stress. Oxidative stress promotes cardiac ryanodine receptor (RyR2) intersubunit crosslinking. Human RyR2 crosslinking promotes Ca leak and calmodulin dissociation. RyR2 C3602 is not involved in crosslinking, slightly affects calmodulin binding. RyR2 crosslinking is an important pathology related RyR2 regulator.
Collapse
Affiliation(s)
- Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago, IL, USA
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, IL, USA
| | - Robyn T Rebbeck
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Daniel Kahn
- Department of Cell and Molecular Physiology, Loyola University Chicago, IL, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, IL, USA.
| |
Collapse
|
102
|
Bauerová-Hlinková V, Hajdúchová D, Bauer JA. Structure and Function of the Human Ryanodine Receptors and Their Association with Myopathies-Present State, Challenges, and Perspectives. Molecules 2020; 25:molecules25184040. [PMID: 32899693 PMCID: PMC7570887 DOI: 10.3390/molecules25184040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 01/28/2023] Open
Abstract
Cardiac arrhythmias are serious, life-threatening diseases associated with the dysregulation of Ca2+ influx into the cytoplasm of cardiomyocytes. This dysregulation often arises from dysfunction of ryanodine receptor 2 (RyR2), the principal Ca2+ release channel. Dysfunction of RyR1, the skeletal muscle isoform, also results in less severe, but also potentially life-threatening syndromes. The RYR2 and RYR1 genes have been found to harbor three main mutation “hot spots”, where mutations change the channel structure, its interdomain interface properties, its interactions with its binding partners, or its dynamics. In all cases, the result is a defective release of Ca2+ ions from the sarcoplasmic reticulum into the myocyte cytoplasm. Here, we provide an overview of the most frequent diseases resulting from mutations to RyR1 and RyR2, briefly review some of the recent experimental structural work on these two molecules, detail some of the computational work describing their dynamics, and summarize the known changes to the structure and function of these receptors with particular emphasis on their N-terminal, central, and channel domains.
Collapse
|
103
|
Guo W, Sun B, Estillore JP, Wang R, Chen SRW. The central domain of cardiac ryanodine receptor governs channel activation, regulation, and stability. J Biol Chem 2020; 295:15622-15635. [PMID: 32878990 DOI: 10.1074/jbc.ra120.013512] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/25/2020] [Indexed: 11/06/2022] Open
Abstract
Structural analyses identified the central domain of ryanodine receptor (RyR) as a transducer converting conformational changes in the cytoplasmic platform to the RyR gate. The central domain is also a regulatory hub encompassing the Ca2+-, ATP-, and caffeine-binding sites. However, the role of the central domain in RyR activation and regulation has yet to be defined. Here, we mutated five residues that form the Ca2+ activation site and 10 residues with negatively charged or oxygen-containing side chains near the Ca2+ activation site. We also generated eight disease-associated mutations within the central domain of RyR2. We determined the effect of these mutations on Ca2+, ATP, and caffeine activation and Mg2+ inhibition of RyR2. Mutating the Ca2+ activation site markedly reduced the sensitivity of RyR2 to Ca2+ and caffeine activation. Unexpectedly, Ca2+ activation site mutation E3848A substantially enhanced the Ca2+-independent basal activity of RyR2, suggesting that E3848A may also affect the stability of the closed state of RyR2. Mutations in the Ca2+ activation site also abolished the effect of ATP/caffeine on the Ca2+-independent basal activity, suggesting that the Ca2+ activation site is also a critical determinant of ATP/caffeine action. Mutating residues with negatively charged or oxygen-containing side chains near the Ca2+ activation site significantly altered Ca2+ and caffeine activation and reduced Mg2+ inhibition. Furthermore, disease-associated RyR2 mutations within the central domain significantly enhanced Ca2+ and caffeine activation and reduced Mg2+ inhibition. Our data demonstrate that the central domain plays an important role in channel activation, channel regulation, and closed state stability.
Collapse
Affiliation(s)
- Wenting Guo
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Bo Sun
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Medical School, Kunming University of Science and Technology, Kunming, China
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ruiwu Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
104
|
Zhang XH, Morad M. Ca 2+ signaling of human pluripotent stem cells-derived cardiomyocytes as compared to adult mammalian cardiomyocytes. Cell Calcium 2020; 90:102244. [PMID: 32585508 PMCID: PMC7483365 DOI: 10.1016/j.ceca.2020.102244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) have been extensively used for in vitro modeling of human cardiovascular disease, drug screening and pharmacotherapy, but little rigorous studies have been reported on their biophysical or Ca2+ signaling properties. There is also considerable concern as to the level of their maturity and whether they can serve as reliable models for adult human cardiac myocytes. Ultrastructural difference such as lack of t-tubular network, their polygonal shapes, disorganized sarcomeric myofilament, and their rhythmic automaticity, among others, have been cited as evidence for immaturity of hiPSC-CMs. In this review, we will deal with Ca2+ signaling, its regulation, and its stage of maturity as compared to the mammalian adult cardiomyocytes. We shall summarize the data on functional aspects of Ca2+signaling and its parameters that include: L-type calcium channel (Cav1.2), ICa-induced Ca2+release, CICR, and its parameters, cardiac Na/Ca exchanger (NCX1), the ryanodine receptors (RyR2), sarco-reticular Ca2+pump, SERCA2a/PLB, and the contribution of mitochondrial Ca2+ to hiPSC-CMs excitation-contraction (EC)-coupling as compared with adult mammalian cardiomyocytes. The comparative studies suggest that qualitatively hiPSC-CMs have similar Ca2+signaling properties as those of adult cardiomyocytes, but quantitative differences do exist. This review, we hope, will allow the readers to judge for themselves to what extent Ca2+signaling of hiPSC-CMs represents the adult form of this signaling pathway, and whether these cells can be used as good models of human cardiomyocytes.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States
| | - Martin Morad
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States.
| |
Collapse
|
105
|
Structural basis for diamide modulation of ryanodine receptor. Nat Chem Biol 2020; 16:1246-1254. [DOI: 10.1038/s41589-020-0627-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022]
|
106
|
Zheng W, Wen H. Investigating dual Ca 2+ modulation of the ryanodine receptor 1 by molecular dynamics simulation. Proteins 2020; 88:1528-1539. [PMID: 32557910 DOI: 10.1002/prot.25971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 05/26/2020] [Accepted: 06/14/2020] [Indexed: 11/09/2022]
Abstract
The ryanodine receptors (RyR) are essential to calcium signaling in striated muscles. A deep understanding of the complex Ca2+ -activation/inhibition mechanism of RyRs requires detailed structural and dynamic information for RyRs in different functional states (eg, with Ca2+ bound to activating or inhibitory sites). Recently, high-resolution structures of the RyR isoform 1 (RyR1) were solved by cryo-electron microscopy, revealing the location of a Ca2+ binding site for activation. Toward elucidating the Ca2+ -modulation mechanism of RyR1, we performed extensive molecular dynamics simulation of the core RyR1 structure in the presence and absence of activating and solvent Ca2+ (total simulation time is >5 μs). In the presence of solvent Ca2+ , Ca2+ binding to the activating site enhanced dynamics of RyR1 with higher inter-subunit flexibility, asymmetric inter-subunit motions, outward domain motions and partial pore dilation, which may prime RyR1 for subsequent channel opening. In contrast, the solvent Ca2+ alone reduced dynamics of RyR1 and led to inward domain motions and pore contraction, which may cause inhibition. Combining our simulation with the map of disease mutation sites in RyR1, we constructed a wiring diagram of key domains coupled via specific hydrogen bonds involving the mutation sites, some of which were modulated by Ca2+ binding. The structural and dynamic information gained from this study will inform future mutational and functional studies of RyR1 activation and inhibition by Ca2+ .
Collapse
Affiliation(s)
- Wenjun Zheng
- Department of Physics, University at Buffalo, Buffalo, New York, USA
| | - Han Wen
- Department of Physics, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
107
|
Kushnir A, Todd JJ, Witherspoon JW, Yuan Q, Reiken S, Lin H, Munce RH, Wajsberg B, Melville Z, Clarke OB, Wedderburn-Pugh K, Wronska A, Razaqyar MS, Chrismer IC, Shelton MO, Mankodi A, Grunseich C, Tarnopolsky MA, Tanji K, Hirano M, Riazi S, Kraeva N, Voermans NC, Gruber A, Allen C, Meilleur KG, Marks AR. Intracellular calcium leak as a therapeutic target for RYR1-related myopathies. Acta Neuropathol 2020; 139:1089-1104. [PMID: 32236737 DOI: 10.1007/s00401-020-02150-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 01/14/2023]
Abstract
RYR1 encodes the type 1 ryanodine receptor, an intracellular calcium release channel (RyR1) on the skeletal muscle sarcoplasmic reticulum (SR). Pathogenic RYR1 variations can destabilize RyR1 leading to calcium leak causing oxidative overload and myopathy. However, the effect of RyR1 leak has not been established in individuals with RYR1-related myopathies (RYR1-RM), a broad spectrum of rare neuromuscular disorders. We sought to determine whether RYR1-RM affected individuals exhibit pathologic, leaky RyR1 and whether variant location in the channel structure can predict pathogenicity. Skeletal muscle biopsies were obtained from 17 individuals with RYR1-RM. Mutant RyR1 from these individuals exhibited pathologic SR calcium leak and increased activity of calcium-activated proteases. The increased calcium leak and protease activity were normalized by ex-vivo treatment with S107, a RyR stabilizing Rycal molecule. Using the cryo-EM structure of RyR1 and a new dataset of > 2200 suspected RYR1-RM affected individuals we developed a method for assigning pathogenicity probabilities to RYR1 variants based on 3D co-localization of known pathogenic variants. This study provides the rationale for a clinical trial testing Rycals in RYR1-RM affected individuals and introduces a predictive tool for investigating the pathogenicity of RYR1 variants of uncertain significance.
Collapse
Affiliation(s)
- Alexander Kushnir
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua J Todd
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Jessica W Witherspoon
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Harvey Lin
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ross H Munce
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Benjamin Wajsberg
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Zephan Melville
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Oliver B Clarke
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kaylee Wedderburn-Pugh
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Muslima S Razaqyar
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Irene C Chrismer
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Monique O Shelton
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Ami Mankodi
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Kurenai Tanji
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michio Hirano
- Department of Neurology, H. Houston Merritt Neuromuscular Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sheila Riazi
- Department of Anesthesia, University of Toronto and Malignant Hyperthermia Investigation Unit, Toronto General Hospital, Toronto, Ontario, Canada
| | - Natalia Kraeva
- Department of Anesthesia, University of Toronto and Malignant Hyperthermia Investigation Unit, Toronto General Hospital, Toronto, Ontario, Canada
| | - Nicol C Voermans
- Department of Neurology, Donders Centre for Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Carolyn Allen
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Katherine G Meilleur
- Neuromuscular Symptoms Unit, Tissue Injury Branch, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA.
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
108
|
Phillips KF, Santos E, Blair RE, Deshpande LS. Targeting Intracellular Calcium Stores Alleviates Neurological Morbidities in a DFP-Based Rat Model of Gulf War Illness. Toxicol Sci 2020; 169:567-578. [PMID: 30859209 PMCID: PMC6542335 DOI: 10.1093/toxsci/kfz070] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gulf War Illness (GWI) is a chronic multi-symptom disorder afflicting the veterans of the First Gulf War, and includes neurological symptoms characterized by depression and memory deficits. Chronic exposure to organophosphates (OPs) is considered a leading cause for GWI, yet its pathobiology is not fully understood. We recently observed chronic elevations in neuronal Ca2+ levels ([Ca2+]i) in an OP-diisopropyl fluorophosphate (DFP)-based rat model for GWI. This study was aimed at identifying mechanisms underlying elevated [Ca2+]i in this DFP model and investigating whether their therapeutic targeting could improve GWI-like neurological morbidities. Male Sprague-Dawley rats (9 weeks) were exposed to DFP (0.5 mg/kg, s.c., 1×-daily for 5 days) and at 3 months postDFP exposure, behavior was assessed and rats were euthanized for protein estimations and ratiometric Fura-2 [Ca2+]i estimations in acutely dissociated hippocampal neurons. In DFP rats, a sustained elevation in intracellular Ca2+ levels occurred, and pharmacological blockade of Ca2+-induced Ca2+-release mechanisms significantly lowered elevated [Ca2+]i in DFP neurons. Significant reductions in the protein levels of the ryanodine receptor (RyR) stabilizing protein Calstabin2 were also noted. Such a posttranslational modification would render RyR “leaky” resulting in sustained DFP [Ca2+]i elevations. Antagonism of RyR with levetiracetam significantly lower elevated [Ca2+]i in DFP neurons and improved GWI-like behavioral symptoms. Since Ca2+ is a major second messenger molecule, such chronic increases in its levels could underlie pathological synaptic plasticity that expresses itself as GWI morbidities. Our studies show that treatment with drugs targeted at blocking intracellular Ca2+ release could be effective therapies for GWI neurological morbidities.
Collapse
Affiliation(s)
| | | | | | - Laxmikant S Deshpande
- Departments of Neurology.,Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298
| |
Collapse
|
109
|
Lin L, Hao Z, Cao P, Yuchi Z. Homology modeling and docking study of diamondback moth ryanodine receptor reveals the mechanisms for channel activation, insecticide binding and resistance. PEST MANAGEMENT SCIENCE 2020; 76:1291-1303. [PMID: 31595631 DOI: 10.1002/ps.5640] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Diamide insecticides, including phthalic and anthranilic diamides, target insect ryanodine receptors (RyRs) and cause misregulation of calcium signaling in insect muscles and neurons. Several resistance mutations have been reported to reduce the efficacy of the diamides, but the exact binding sites and mechanism of resistance mutations are not clear. RESULTS The recent breakthrough in structural studies of mammalian RyRs has deepened our understanding of these giant calcium-release channels, but structural information about insect RyRs is still scarce. The only reported high-resolution structure is from the N-terminal domain of diamondback moth (DBM) RyR determined by our group. Here, we generate several homology models of full-length DBM RyR representing different functional states and dock the diamide insecticides into the structural models using Schrodinger software. These models reveal the specific structural features, activation mechanism, structural difference between functional states, ligand-binding sites and insecticide-binding sites of DBM RyR. By comparing the structures of wild-type and insecticide-resistant mutants, we propose a model depicting how the mutations affect the insecticide binding. We also identify the key difference between mammalian and insect RyRs that may explain the species-specific binding properties of diamides. CONCLUSION The binding sites for three activators Ca2+ , ATP and caffeine, and regulator ryanodine are conserved in insect and mammalian RyRs, but the binding site for diamide insecticides is species-specific. The phthalic and anthranilic diamides have distinct binding properties in DBM, which can be interfered by resistance mutations located in the transmembrane region. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lianyun Lin
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Zhiyuan Hao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Peng Cao
- Key Laboratory of Drug Targets and Drug Leads for Degenerative Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiguang Yuchi
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
110
|
Chen W, Kudryashev M. Structure of RyR1 in native membranes. EMBO Rep 2020; 21:e49891. [PMID: 32147968 PMCID: PMC7202208 DOI: 10.15252/embr.201949891] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 12/27/2022] Open
Abstract
Ryanodine receptor 1 (RyR1) mediates excitation–contraction coupling by releasing Ca2+ from sarcoplasmic reticulum (SR) to the cytoplasm of skeletal muscle cells. RyR1 activation is regulated by several proteins from both the cytoplasm and lumen of the SR. Here, we report the structure of RyR1 from native SR membranes in closed and open states. Compared to the previously reported structures of purified RyR1, our structure reveals helix‐like densities traversing the bilayer approximately 5 nm from the RyR1 transmembrane domain and sarcoplasmic extensions linking RyR1 to a putative calsequestrin network. We document the primary conformation of RyR1 in situ and its structural variations. The activation of RyR1 is associated with changes in membrane curvature and movement in the sarcoplasmic extensions. Our results provide structural insight into the mechanism of RyR1 in its native environment.
Collapse
Affiliation(s)
- Wenbo Chen
- Max Planck Institute for Biophysics, Frankfurt on Main, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt on Main, Germany
| | - Mikhail Kudryashev
- Max Planck Institute for Biophysics, Frankfurt on Main, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt on Main, Germany
| |
Collapse
|
111
|
Graham B, Shaw MA, Hope IA. Single Amino Acid Changes in the Ryanodine Receptor in the Human Population Have Effects In Vivo on Caenorhabditis elegans Neuro-Muscular Function. Front Genet 2020; 11:37. [PMID: 32174957 PMCID: PMC7054344 DOI: 10.3389/fgene.2020.00037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/13/2020] [Indexed: 01/21/2023] Open
Abstract
The ryanodine receptor mediates intracellular calcium ion release with excitation of nerve and muscle cells. Ryanodine receptor missense variants cause a number of myopathologies, such as malignant hyperthermia, and have been linked with various neuropathologies, including Alzheimer's disease. We characterized the consequences of ryanodine receptor variants in vivo. Eight Caenorhabditis elegans strains, with ryanodine receptor modifications equivalent to human myopathic RYR1 variants, were generated by genome editing. In humans, these variants are rare and confer sensitivity to the inhalational anaesthetic halothane when heterozygous. Increased sensitivity to halothane was found in both homozygous and heterozygous C. elegans. Close analysis revealed distinct subtle locomotion defects, due to the different single amino acid residue changes, even in the absence of the external triggering agent. Distinct pre- and postsynaptic consequences of the variants were characterized through the responses to cholinergic pharmacological agents. The range of phenotypes reflects the complexity of the regulatory inputs to the ryanodine receptor and the criticality of the calcium ion channel opening properties, in different cell types and with age. Ryanodine receptors with these single amino acid residue changes still function as calcium ion channels, but with altered properties which are likely to have subtle consequences for human carriers of such variants. The long-term consequences of subtly altered calcium ion signalling could be cumulative and may be focussed in the smaller nerve cells rather than the more robust muscle cells. It was important to assess phenotypes in vivo to properly appreciate consequences for a whole organism.
Collapse
Affiliation(s)
- Brittany Graham
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Marie-Anne Shaw
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds, United Kingdom
| | - Ian A. Hope
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
112
|
Shimomura T, Yonekawa Y, Nagura H, Tateyama M, Fujiyoshi Y, Irie K. A native prokaryotic voltage-dependent calcium channel with a novel selectivity filter sequence. eLife 2020; 9:52828. [PMID: 32093827 PMCID: PMC7041947 DOI: 10.7554/elife.52828] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/07/2020] [Indexed: 12/20/2022] Open
Abstract
Voltage-dependent Ca2+ channels (Cavs) are indispensable for coupling action potentials with Ca2+ signaling in living organisms. The structure of Cavs is similar to that of voltage-dependent Na+ channels (Navs). It is known that prokaryotic Navs can obtain Ca2+ selectivity by negative charge mutations of the selectivity filter, but native prokaryotic Cavs had not yet been identified. We report the first identification of a native prokaryotic Cav, CavMr, whose selectivity filter contains a smaller number of negatively charged residues than that of artificial prokaryotic Cavs. A relative mutant whose selectivity filter was replaced with that of CavMr exhibits high Ca2+ selectivity. Mutational analyses revealed that the glycine residue of the CavMr selectivity filter is a determinant for Ca2+ selectivity. This glycine residue is well conserved among subdomains I and III of eukaryotic Cavs. These findings provide new insight into the Ca2+ selectivity mechanism that is conserved from prokaryotes to eukaryotes. Electrical signals in the brain and muscles allow animals – including humans – to think, make memories and move around. Cells generate these signals by enabling charged particles known as ions to pass through the physical barrier that surrounds all cells, the cell membrane, at certain times and in certain locations. The ions pass through pores made by various channel proteins, which generally have so-called “selectivity filters” that only allow particular types of ions to fit through. For example, the selectivity filters of a family of channels in mammals known as the Cavs only allow calcium ions to pass through. Another family of ion channels in mammals are similar in structure to the Cavs but their selectivity filters only allow sodium ions to pass through instead of calcium ions. Ion channels are found in all living cells including in bacteria. It is thought that the Cavs and sodium-selective channels may have both evolved from Cav-like channels in an ancient lifeform that was the common ancestor of modern bacteria and animals. Previous studies in bacteria found that modifying the selectivity filters of some sodium-selective channels known as BacNavs allowed calcium ions to pass through the mutant channels instead of sodium ions. However, no Cav channels had been identified in bacteria so far, representing a missing link in the evolutionary history of ion channels. Shimomura et al. have now found a Cav-like channel in a bacterium known as Meiothermus ruber. Like all proteins, ion channels are made from amino acids and comparing the selectivity filter of the M. ruber Cav with those of mammalian Cavs and the calcium-selective BacNav mutants from previous studies revealed one amino acid that plays a particularly important role. This amino acid is a glycine that helps select which ions may pass through the pore and is also present in the selectivity filters of many Cavs in mammals. Together these findings suggest that the Cav channel from M. ruber is similar to the mammal Cav channels and may more closely resemble the Cav-like channels thought to have existed in the common ancestor of bacteria and animals. Since other channel proteins from bacteria are useful genetic tools for studies in human and other animal cells, the Cav channel from M. ruber has the potential to be used to stimulate calcium signaling in experiments.
Collapse
Affiliation(s)
- Takushi Shimomura
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Nagoya, Japan.,Division of Biophysics and Neurobiology, National Institute for Physiological Sciences, Okazaki, Japan
| | - Yoshiki Yonekawa
- Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Hitoshi Nagura
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Nagoya, Japan
| | - Michihiro Tateyama
- Division of Biophysics and Neurobiology, National Institute for Physiological Sciences, Okazaki, Japan
| | - Yoshinori Fujiyoshi
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Nagoya, Japan.,CeSPIA Inc, Tokyo, Japan
| | - Katsumasa Irie
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Nagoya, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
113
|
Seidel M, de Meritens CR, Johnson L, Parthimos D, Bannister M, Thomas NL, Ozekhome-Mike E, Lai FA, Zissimopoulos S. Identification of an amino-terminus determinant critical for ryanodine receptor/Ca2+ release channel function. Cardiovasc Res 2020; 117:780-791. [PMID: 32077934 PMCID: PMC7898959 DOI: 10.1093/cvr/cvaa043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/05/2019] [Accepted: 02/17/2020] [Indexed: 01/12/2023] Open
Abstract
AIMS The cardiac ryanodine receptor (RyR2), which mediates intracellular Ca2+ release to trigger cardiomyocyte contraction, participates in development of acquired and inherited arrhythmogenic cardiac disease. This study was undertaken to characterize the network of inter- and intra-subunit interactions regulating the activity of the RyR2 homotetramer. METHODS AND RESULTS We use mutational investigations combined with biochemical assays to identify the peptide sequence bridging the β8 with β9 strand as the primary determinant mediating RyR2 N-terminus self-association. The negatively charged side chains of two aspartate residues (D179 and D180) within the β8-β9 loop are crucial for the N-terminal inter-subunit interaction. We also show that the RyR2 N-terminus domain interacts with the C-terminal channel pore region in a Ca2+-independent manner. The β8-β9 loop is required for efficient RyR2 subunit oligomerization but it is dispensable for N-terminus interaction with C-terminus. Deletion of the β8-β9 sequence produces unstable tetrameric channels with subdued intracellular Ca2+ mobilization implicating a role for this domain in channel opening. The arrhythmia-linked R176Q mutation within the β8-β9 loop decreases N-terminus tetramerization but does not affect RyR2 subunit tetramerization or the N-terminus interaction with C-terminus. RyR2R176Q is a characteristic hypersensitive channel displaying enhanced intracellular Ca2+ mobilization suggesting an additional role for the β8-β9 domain in channel closing. CONCLUSION These results suggest that efficient N-terminus inter-subunit communication mediated by the β8-β9 loop may constitute a primary regulatory mechanism for both RyR2 channel activation and suppression.
Collapse
Affiliation(s)
- Monika Seidel
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Swansea University Medical School, Institute of Life Science, Swansea SA2 8PP, UK
| | - Camille Rabesahala de Meritens
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Swansea University Medical School, Institute of Life Science, Swansea SA2 8PP, UK
| | - Louisa Johnson
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Swansea University Medical School, Institute of Life Science, Swansea SA2 8PP, UK
| | - Dimitris Parthimos
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Mark Bannister
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Swansea University Medical School, Institute of Life Science, Swansea SA2 8PP, UK
| | - Nia Lowri Thomas
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,School of Pharmacy & Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
| | - Esizaze Ozekhome-Mike
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Swansea University Medical School, Institute of Life Science, Swansea SA2 8PP, UK
| | - Francis Anthony Lai
- College of Medicine, QU Health, and Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Spyros Zissimopoulos
- Department of Cardiology, School of Medicine, Wales Heart Research Institute, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.,Swansea University Medical School, Institute of Life Science, Swansea SA2 8PP, UK
| |
Collapse
|
114
|
Calmodulin Mutations Associated with Heart Arrhythmia: A Status Report. Int J Mol Sci 2020; 21:ijms21041418. [PMID: 32093079 PMCID: PMC7073091 DOI: 10.3390/ijms21041418] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Calmodulin (CaM) is a ubiquitous intracellular Ca2+ sensing protein that modifies gating of numerous ion channels. CaM has an extraordinarily high level of evolutionary conservation, which led to the fundamental assumption that mutation would be lethal. However, in 2012, complete exome sequencing of infants suffering from recurrent cardiac arrest revealed de novo mutations in the three human CALM genes. The correlation between mutations and pathophysiology suggests defects in CaM-dependent ion channel functions. Here, we review the current state of the field for all reported CaM mutations associated with cardiac arrhythmias, including knowledge of their biochemical and structural characteristics, and progress towards understanding how these mutations affect cardiac ion channel function.
Collapse
|
115
|
Schartner V, Laporte J, Böhm J. Abnormal Excitation-Contraction Coupling and Calcium Homeostasis in Myopathies and Cardiomyopathies. J Neuromuscul Dis 2020; 6:289-305. [PMID: 31356215 DOI: 10.3233/jnd-180314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Muscle contraction requires specialized membrane structures with precise geometry and relies on the concerted interplay of electrical stimulation and Ca2+ release, known as excitation-contraction coupling (ECC). The membrane structure hosting ECC is called triad in skeletal muscle and dyad in cardiac muscle, and structural or functional defects of triads and dyads have been observed in a variety of myopathies and cardiomyopathies. Based on their function, the proteins localized at the triad/dyad can be classified into three molecular pathways: the Ca2+ release complex (CRC), store-operated Ca2+ entry (SOCE), and membrane remodeling. All three are mechanistically linked, and consequently, aberrations in any of these pathways cause similar disease entities. This review provides an overview of the clinical and genetic spectrum of triad and dyad defects with a main focus of attention on the underlying pathomechanisms.
Collapse
Affiliation(s)
- Vanessa Schartner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| |
Collapse
|
116
|
Zhou Y, Ma D, Lin L, You M, Yuchi Z, You S. Crystal Structure of the Ryanodine Receptor SPRY2 Domain from the Diamondback Moth Provides Insights into the Development of Novel Insecticides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1731-1740. [PMID: 31951399 DOI: 10.1021/acs.jafc.9b08151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Diamide insecticides targeting ryanodine receptors (RyRs) are a major class of pesticides used to control a wide range of agricultural pests, but their efficacies have been reduced dramatically by the recent emergence of resistance mutations. There is a pressing need to develop novel insecticides, targeting distinct and novel binding sites within insect RyRs to overcome the resistance crisis; however, the limited structural information on insect RyRs is a major roadblock to our understanding of their molecular mechanisms. Here, we report the crystal structure of the RyR SPRY2 domain from the diamondback moth (DBM), Plutella xylostella, a destructive agricultural pest worldwide that has developed resistance to all classes of insecticide at 2.06 Å resolution. The overall fold of DBM SPRY2 is similar to its mammalian homolog, but it shows distinct conformations in several loops. Docking it into the recently published cryo-electron microscope structure of the full-length RyR reveals that two insect-specific loops interact with the BSol domain from the neighboring subunit. The SPRY2-BSol interface will change the conformation upon channel gating, indicating that it might be a potential targeting site for insect-specific insecticides. Interestingly, several previously identified disease-causing mutations also lie in the same interface, implying that this interface is important for channel gating. Another insect-specific loop located in the SPRY2-SPRY3 interface might indirectly affect another gating interface between SPRY3 and Repeat34.
Collapse
Affiliation(s)
- Yuanyuan Zhou
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology , Fujian Agriculture and Forestry University , Fuzhou 350002 , China
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology , Tianjin University , Tianjin 300072 , China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education , Fuzhou 350002 , China
- Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture , Fuzhou 350002 , China
| | - Dan Ma
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology , Tianjin University , Tianjin 300072 , China
| | - Lianyun Lin
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology , Tianjin University , Tianjin 300072 , China
| | - Minsheng You
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology , Fujian Agriculture and Forestry University , Fuzhou 350002 , China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education , Fuzhou 350002 , China
- Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture , Fuzhou 350002 , China
| | - Zhiguang Yuchi
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology , Fujian Agriculture and Forestry University , Fuzhou 350002 , China
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology , Tianjin University , Tianjin 300072 , China
| | - Shijun You
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology , Fujian Agriculture and Forestry University , Fuzhou 350002 , China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education , Fuzhou 350002 , China
- Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture , Fuzhou 350002 , China
| |
Collapse
|
117
|
Ogawa H, Kurebayashi N, Yamazawa T, Murayama T. Regulatory mechanisms of ryanodine receptor/Ca 2+ release channel revealed by recent advancements in structural studies. J Muscle Res Cell Motil 2020; 42:291-304. [PMID: 32040690 PMCID: PMC8332584 DOI: 10.1007/s10974-020-09575-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
Ryanodine receptors (RyRs) are huge homotetrameric Ca2+ release channels localized to the sarcoplasmic reticulum. RyRs are responsible for the release of Ca2+ from the SR during excitation–contraction coupling in striated muscle cells. Recent revolutionary advancements in cryo-electron microscopy have provided a number of near-atomic structures of RyRs, which have enabled us to better understand the architecture of RyRs. Thus, we are now in a new era understanding the gating, regulatory and disease-causing mechanisms of RyRs. Here we review recent advances in the elucidation of the structures of RyRs, especially RyR1 in skeletal muscle, and their mechanisms of regulation by small molecules, associated proteins and disease-causing mutations.
Collapse
Affiliation(s)
- Haruo Ogawa
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan.
| | - Nagomi Kurebayashi
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Toshiko Yamazawa
- Department of Molecular Physiology, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Takashi Murayama
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| |
Collapse
|
118
|
McCarthy MR, Savich Y, Cornea RL, Thomas DD. Resolved Structural States of Calmodulin in Regulation of Skeletal Muscle Calcium Release. Biophys J 2020; 118:1090-1100. [PMID: 32049056 DOI: 10.1016/j.bpj.2020.01.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 12/22/2022] Open
Abstract
Calmodulin (CaM) is proposed to modulate activity of the skeletal muscle sarcoplasmic reticulum (SR) calcium release channel (ryanodine receptor, RyR1 isoform) via a mechanism dependent on the conformation of RyR1-bound CaM. However, the correlation between CaM structure and functional regulation of RyR in physiologically relevant conditions is largely unknown. Here, we have used time-resolved fluorescence resonance energy transfer (TR-FRET) to study structural changes in CaM that may play a role in the regulation of RyR1. We covalently labeled each lobe of CaM (N and C) with fluorescent probes and used intramolecular TR-FRET to assess interlobe distances when CaM is bound to RyR1 in SR membranes, purified RyR1, or a peptide corresponding to the CaM-binding domain of RyR (RyRp). TR-FRET resolved an equilibrium between two distinct structural states (conformations) of CaM, each characterized by an interlobe distance and Gaussian distribution width (disorder). In isolated CaM, at low Ca2+, the two conformations of CaM are resolved, centered at 5 nm (closed) and 7 nm (open). At high Ca2+, the equilibrium shifts to favor the open conformation. In the presence of RyRp at high Ca2+, the closed conformation shifts to a more compact conformation and is the major component. When CaM is bound to full-length RyR1, either purified or in SR membranes, strikingly different results were obtained: 1) the two conformations are resolved and more ordered, 2) the open state is the major component, and 3) Ca2+ stabilized the closed conformation by a factor of two. We conclude that the Ca2+-dependent structural distribution of CaM bound to RyR1 is distinct from that of CaM bound to RyRp. We propose that the function of RyR1 is tuned to the Ca2+-dependent structural dynamics of bound CaM.
Collapse
Affiliation(s)
- Megan R McCarthy
- Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, Minnesota
| | - Yahor Savich
- Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, Minnesota; School of Physics and Astronomy, University of Minnesota, Minneapolis, Minnesota
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, Minnesota
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, Minnesota.
| |
Collapse
|
119
|
Shishmarev D. Excitation-contraction coupling in skeletal muscle: recent progress and unanswered questions. Biophys Rev 2020; 12:143-153. [PMID: 31950344 DOI: 10.1007/s12551-020-00610-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Excitation-contraction coupling (ECC) is a physiological process that links excitation of muscles by the nervous system to their mechanical contraction. In skeletal muscle, ECC is initiated with an action potential, generated by the somatic nervous system, which causes a depolarisation of the muscle fibre membrane (sarcolemma). This leads to a rapid change in the transmembrane potential, which is detected by the voltage-gated Ca2+ channel dihydropyridine receptor (DHPR) embedded in the sarcolemma. DHPR transmits the contractile signal to another Ca2+ channel, ryanodine receptor (RyR1), embedded in the membrane of the sarcoplasmic reticulum (SR), which releases a large amount of Ca2+ ions from the SR that initiate muscle contraction. Despite the fundamental role of ECC in skeletal muscle function of all vertebrate species, the molecular mechanism underpinning the communication between the two key proteins involved in the process (DHPR and RyR1) is still largely unknown. The goal of this work is to review the recent progress in our understanding of ECC in skeletal muscle from the point of view of the structure and interactions of proteins involved in the process, and to highlight the unanswered questions in the field.
Collapse
Affiliation(s)
- Dmitry Shishmarev
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
120
|
Connell P, Word TA, Wehrens XHT. Targeting pathological leak of ryanodine receptors: preclinical progress and the potential impact on treatments for cardiac arrhythmias and heart failure. Expert Opin Ther Targets 2020; 24:25-36. [PMID: 31869254 DOI: 10.1080/14728222.2020.1708326] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction: Type-2 ryanodine receptor (RyR2) located on the sarcoplasmic reticulum initiate systolic Ca2+ transients within cardiomyocytes. Proper functioning of RyR2 is therefore crucial to the timing and force generated by cardiomyocytes within a healthy heart. Improper intracellular Ca2+ handing secondary to RyR2 dysfunction is associated with a variety of cardiac pathologies including catecholaminergic polymorphic ventricular tachycardia (CPVT), atrial fibrillation (AF), and heart failure (HF). Thus, RyR2 and its associated accessory proteins provide promising drug targets to scientists developing therapeutics for a variety of cardiac pathologies.Areas covered: In this article, we review the role of RyR2 in a variety of cardiac pathologies. We performed a literature search utilizing PubMed and MEDLINE as well as reviewed registries of trials from clinicaltrials.gov from 2010 to 2019 for novel therapeutic approaches that address the cellular mechanisms underlying CPVT, AF, and HF by specifically targeting defective RyR2 channels.Expert opinion: The negative impact of cardiac dysfunction on human health and medical economics are major motivating factors for establishing new and effective therapeutic approaches. Focusing on directly impacting the molecular mechanisms underlying defective Ca2+ handling by RyR2 in HF and arrhythmia has great potential to be translated into novel and innovative therapies.
Collapse
Affiliation(s)
- Patrick Connell
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.,Departments of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Tarah A Word
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.,Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.,Departments of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA.,Medicine (Cardiology, Baylor College of Medicine, Houston, TX, USA.,Neuroscience, Baylor College of Medicine, Houston, TX, USA.,Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
121
|
Yousuf MS, Maguire AD, Simmen T, Kerr BJ. Endoplasmic reticulum-mitochondria interplay in chronic pain: The calcium connection. Mol Pain 2020; 16:1744806920946889. [PMID: 32787562 PMCID: PMC7427143 DOI: 10.1177/1744806920946889] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic pain is a debilitating condition that affects roughly a third to a half of the world's population. Despite its substantial effect on society, treatment for chronic pain is modest, at best, notwithstanding its side effects. Hence, novel therapeutics are direly needed. Emerging evidence suggests that calcium plays an integral role in mediating neuronal plasticity that underlies sensitization observed in chronic pain states. The endoplasmic reticulum and the mitochondria are the largest calcium repositories in a cell. Here, we review how stressors, like accumulation of misfolded proteins and oxidative stress, influence endoplasmic reticulum and mitochondria function and contribute to chronic pain. We further examine the shuttling of calcium across the mitochondrial-associated membrane as a mechanism of cross-talk between the endoplasmic reticulum and the mitochondria. In addition, we discuss how endoplasmic reticulum stress, mitochondrial impairment, and calcium dyshomeostasis are implicated in various models of neuropathic pain. We propose a novel framework of endoplasmic reticulum-mitochondria signaling in mediating pain hypersensitivity. These observations require further investigation in order to develop novel therapies for chronic pain.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Aislinn D Maguire
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Thomas Simmen
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Pharmacology, University of Alberta, Edmonton, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
122
|
Tran QK. Reciprocality Between Estrogen Biology and Calcium Signaling in the Cardiovascular System. Front Endocrinol (Lausanne) 2020; 11:568203. [PMID: 33133016 PMCID: PMC7550652 DOI: 10.3389/fendo.2020.568203] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/19/2020] [Indexed: 12/30/2022] Open
Abstract
17β-Estradiol (E2) is the main estrogenic hormone in the body and exerts many cardiovascular protective effects. Via three receptors known to date, including estrogen receptors α (ERα) and β (ERβ) and the G protein-coupled estrogen receptor 1 (GPER, aka GPR30), E2 regulates numerous calcium-dependent activities in cardiovascular tissues. Nevertheless, effects of E2 and its receptors on components of the calcium signaling machinery (CSM), the underlying mechanisms, and the linked functional impact are only beginning to be elucidated. A picture is emerging of the reciprocality between estrogen biology and Ca2+ signaling. Therein, E2 and GPER, via both E2-dependent and E2-independent actions, moderate Ca2+-dependent activities; in turn, ERα and GPER are regulated by Ca2+ at the receptor level and downstream signaling via a feedforward loop. This article reviews current understanding of the effects of E2 and its receptors on the cardiovascular CSM and vice versa with a focus on mechanisms and combined functional impact. An overview of the main CSM components in cardiovascular tissues will be first provided, followed by a brief review of estrogen receptors and their Ca2+-dependent regulation. The effects of estrogenic agonists to stimulate acute Ca2+ signals will then be reviewed. Subsequently, E2-dependent and E2-independent effects of GPER on components of the Ca2+ signals triggered by other stimuli will be discussed. Finally, a case study will illustrate how the many mechanisms are coordinated to moderate Ca2+-dependent activities in the cardiovascular system.
Collapse
|
123
|
Marques F, Saro G, Lia AS, Poole RJ, Falquet L, Glauser DA. Identification of avoidance genes through neural pathway-specific forward optogenetics. PLoS Genet 2019; 15:e1008509. [PMID: 31891575 PMCID: PMC6938339 DOI: 10.1371/journal.pgen.1008509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/05/2019] [Indexed: 11/19/2022] Open
Abstract
Understanding how the nervous system bridges sensation and behavior requires the elucidation of complex neural and molecular networks. Forward genetic approaches, such as screens conducted in C. elegans, have successfully identified genes required to process natural sensory stimuli. However, functional redundancy within the underlying neural circuits, which are often organized with multiple parallel neural pathways, limits our ability to identify 'neural pathway-specific genes', i.e. genes that are essential for the function of some, but not all of these redundant neural pathways. To overcome this limitation, we developed a 'forward optogenetics' screening strategy in which natural stimuli are initially replaced by the selective optogenetic activation of a specific neural pathway. We used this strategy to address the function of the polymodal FLP nociceptors mediating avoidance of noxious thermal and mechanical stimuli. According to our expectations, we identified both mutations in 'general' avoidance genes that broadly impact avoidance responses to a variety of natural noxious stimuli (unc-4, unc-83, and eat-4) and mutations that produce a narrower impact, more restricted to the FLP pathway (syd-2, unc-14 and unc-68). Through a detailed follow-up analysis, we further showed that the Ryanodine receptor UNC-68 acts cell-autonomously in FLP to adjust heat-evoked calcium signals and aversive behaviors. As a whole, our work (i) reveals the importance of properly regulated ER calcium release for FLP function, (ii) provides new entry points for new nociception research and (iii) demonstrates the utility of our forward optogenetic strategy, which can easily be transposed to analyze other neural pathways.
Collapse
Affiliation(s)
- Filipe Marques
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Gabriella Saro
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Andrei-Stefan Lia
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Richard J. Poole
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Swiss Institute of Bioinformatics, Fribourg, Switzerland
| | | |
Collapse
|
124
|
Ca 2+ Channels Mediate Bidirectional Signaling between Sarcolemma and Sarcoplasmic Reticulum in Muscle Cells. Cells 2019; 9:cells9010055. [PMID: 31878335 PMCID: PMC7016941 DOI: 10.3390/cells9010055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
The skeletal muscle and myocardial cells present highly specialized structures; for example, the close interaction between the sarcoplasmic reticulum (SR) and mitochondria—responsible for excitation-metabolism coupling—and the junction that connects the SR with T-tubules, critical for excitation-contraction (EC) coupling. The mechanisms that underlie EC coupling in these two cell types, however, are fundamentally distinct. They involve the differential expression of Ca2+ channel subtypes: CaV1.1 and RyR1 (skeletal), vs. CaV1.2 and RyR2 (cardiac). The CaV channels transform action potentials into elevations of cytosolic Ca2+, by activating RyRs and thus promoting SR Ca2+ release. The high levels of Ca2+, in turn, stimulate not only the contractile machinery but also the generation of mitochondrial reactive oxygen species (ROS). This forward signaling is reciprocally regulated by the following feedback mechanisms: Ca2+-dependent inactivation (of Ca2+ channels), the recruitment of Na+/Ca2+ exchanger activity, and oxidative changes in ion channels and transporters. Here, we summarize both well-established concepts and recent advances that have contributed to a better understanding of the molecular mechanisms involved in this bidirectional signaling.
Collapse
|
125
|
Yamaguchi N. Molecular Insights into Calcium Dependent Regulation of Ryanodine Receptor Calcium Release Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1131:321-336. [DOI: 10.1007/978-3-030-12457-1_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
126
|
Ma Z, Liu H, Yu H. Triclosan Affects Ca 2+ Regulatory Module and Musculature Development in Skeletal Myocyte during Early Life Stages of Zebrafish ( Danio rerio). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:11988-11998. [PMID: 31532625 DOI: 10.1021/acs.est.9b03231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Advanced technologies for toxicity tests are designed to identify biomarkers with superior predictive power or end points of the complex web of biological pathways. However, the data obtained need to be fully characterized for dose-response, physiological systems, and relevance to a system or (sub) population before biological interpretation and decision making. In this study, the toxicity of triclosan (TCS) on zebrafish was selected as a case study to correlate the observed morphological effects with existing data and identify the critical events by receptor activity sensitivity analysis. Triclosan exhibited weak acute toxicity against zebrafish and significantly affected the development of trunk muscles at 0.52, 1.04, and 1.73 μM. Through receptor-mediated screening, we found that the adverse effects of TCS induce Ryanodine receptor 1 (RyR1) activity and distort Ca2+ signaling. The trunk skeletal muscle abnormalities occurred only when the dihydropyridine receptor (DHPR) was blocked, demonstrating that TCS mainly influenced the Ca2+ regulatory module associated with signaling between DHPRs and RyR1; DHPRs mainly regulated the orthograde and retrograde signaling in skeletal muscles. This unexpected result could integrate the mode of action of TCS and provide insight for high-throughput screening and toxicity prediction using zebrafish.
Collapse
Affiliation(s)
- Zhiyuan Ma
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment , Nanjing University , Nanjing 210023 , China
| | - Hongling Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment , Nanjing University , Nanjing 210023 , China
| | - Hongxia Yu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment , Nanjing University , Nanjing 210023 , China
| |
Collapse
|
127
|
Xu T, Yuchi Z. Crystal structure of diamondback moth ryanodine receptor Repeat34 domain reveals insect-specific phosphorylation sites. BMC Biol 2019; 17:77. [PMID: 31597572 PMCID: PMC6784350 DOI: 10.1186/s12915-019-0698-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/02/2019] [Indexed: 01/23/2023] Open
Abstract
Background Ryanodine receptor (RyR), a calcium-release channel located in the sarcoplasmic reticulum membrane of muscles, is the target of insecticides used against a wide range of agricultural pests. Mammalian RyRs have been shown to be under the regulatory control of several kinases and phosphatases, but little is known about the regulation of insect RyRs by phosphorylation. Results Here we present the crystal structures of wild-type and phospho-mimetic RyR Repeat34 domain containing PKA phosphorylation sites from diamondback moth (DBM), a major lepidopteran pest of cruciferous vegetables. The structure has unique features, not seen in mammalian RyRs, including an additional α-helix near the phosphorylation loop. Using tandem mass spectrometry, we identify several PKA sites clustering in the phosphorylation loop and the newly identified α-helix. Bioinformatics analysis shows that this α-helix is only present in Lepidoptera, suggesting an insect-specific regulation. Interestingly, the specific phosphorylation pattern is temperature-dependent. The thermal stability of the DBM Repeat34 domain is significantly lower than that of the analogous domain in the three mammalian RyR isoforms, indicating a more dynamic domain structure that can be partially unfolded to facilitate the temperature-dependent phosphorylation. Docking the structure into the cryo-electron microscopy model of full-length RyR reveals that the interface between the Repeat34 and neighboring HD1 domain is more conserved than that of the phosphorylation loop region that might be involved in the interaction with SPRY3 domain. We also identify an insect-specific glycerol-binding pocket that could be potentially targeted by novel insecticides to fight the current resistance crisis. Conclusions The crystal structures of the DBM Repeat34 domain reveals insect-specific temperature-dependent phosphorylation sites that may regulate insect ryanodine receptor function. It also reveals insect-specific structural features and a potential ligand-binding site that could be targeted in an effort to develop green pesticides with high species-specificity.
Collapse
Affiliation(s)
- Tong Xu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Zhiguang Yuchi
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
128
|
Targeted mutagenesis of the ryanodine receptor by Platinum TALENs causes slow swimming behaviour in Pacific bluefin tuna (Thunnus orientalis). Sci Rep 2019; 9:13871. [PMID: 31554877 PMCID: PMC6761128 DOI: 10.1038/s41598-019-50418-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/05/2019] [Indexed: 11/08/2022] Open
Abstract
In bluefin tuna aquaculture, high mortalities of hatchery-reared juveniles occur in sea cages owing to wall collisions that are caused by high-speed swimming in panic due to changes in illuminance. Here, we report that targeted gene mutagenesis of the ryanodine receptor (RyR1b), which allows the sarcoplasmic reticulum to release Ca2+ in fast skeletal muscle, using highly active Platinum TALENs caused slow swimming behaviour in response to external stimuli in Pacific bluefin tuna (PBT) larvae. This characteristic would be a useful trait to prevent wall collisions in aquaculture production. A pair of Platinum TALENs targeting exons 2 and 43 of the PBT ryr1b gene induced deletions in each TALEN target site of the injected embryos with extremely high efficiency. In addition, ryr1b expression was significantly decreased in the mutated G0 larvae at 7 days after hatching (DAH). A touch-evoked escape behaviour assay revealed that the ryr1b-mutated PBT larvae swam away much less efficiently in response to mechanosensory stimulation at 7 DAH than did the wild-type larvae. Our results demonstrate that genome editing technologies are effective tools for determining the functional characterization of genes in a comparatively short period, and create avenues for facilitating genetic studies and breeding of bluefin tuna species.
Collapse
|
129
|
Diszházi G, Magyar ZÉ, Mótyán JA, Csernoch L, Jóna I, Nánási PP, Almássy J. Dantrolene Requires Mg 2+ and ATP To Inhibit the Ryanodine Receptor. Mol Pharmacol 2019; 96:401-407. [PMID: 31337666 DOI: 10.1124/mol.119.116475] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/07/2019] [Indexed: 12/15/2022] Open
Abstract
Dantrolene is a ryanodine receptor (RyR) inhibitor, which is used to relax muscles in malignant hyperthermia syndrome. Although dantrolene binds to the RyR protein, its mechanism of action is unknown, mainly because of the controversial results showing that dantrolene inhibited Ca2+ release from intact fibers and sarcoplasmic reticulum (SR) vesicles, but failed to inhibit single RyR channel currents in bilayers. Accordingly, it was concluded that an important factor for dantrolene's action was lost during the purification procedure of RyR. Recently, Mg2+ was demonstrated to be the essential factor for dantrolene to inhibit Ca2+ release in skinned muscle fibers. The aim of the present study was to confirm these results in Ca2+ release and bilayer experiments, using SR vesicles and solubilized channels, respectively. Our Ca2+ release experiments demonstrated that the effect of dantrolene and Mg2+ was cooperative and that ATP enhanced the inhibiting effect of dantrolene. Namely, 10 µM dantrolene reduced RyR channel open probability by ∼50% in the presence of 3 mM free Mg2+ and 1 mM ATP, whereas channel activity further decreased to ∼20% of control when [ATP] was increased to 2 mM. Our data provide important complementary information that supports the direct, Mg2+-dependent mechanism of dantrolene's action and suggests that dantrolene also requires ATP to inhibit RyR.
Collapse
Affiliation(s)
- Gyula Diszházi
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Édua Magyar
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| | - János András Mótyán
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| | - László Csernoch
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| | - István Jóna
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| | - Péter Pál Nánási
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| | - János Almássy
- Departments of Physiology (G.D., Z.É.M., L.C., P.P.N., J.A.) and Biochemistry and Molecular Biology (J.A.M.), and Research Centre for Molecular Medicine (I.J.), Faculty of Medicine, and Department of Dental Physiology and Pharmacology, Faculty of Dentistry (P.P.N.), University of Debrecen, Debrecen, Hungary
| |
Collapse
|
130
|
Resveratrol Directly Controls the Activity of Neuronal Ryanodine Receptors at the Single-Channel Level. Mol Neurobiol 2019; 57:422-434. [PMID: 31376069 DOI: 10.1007/s12035-019-01705-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 07/10/2019] [Indexed: 01/14/2023]
Abstract
Calcium ion dyshomeostasis contributes to the progression of many neurodegenerative diseases and represents a target for the development of neuroprotective therapies, as reported by Duncan et al. (Molecules 15(3):1168-95, 2010), LaFerla (Nat Rev Neurosci 3(11):862-72, 2002), and Niittykoshi et al. (Invest Ophthalmol Vis Sci 51(12):6387-93, 2010). Dysfunctional ryanodine receptors contribute to calcium ion dyshomeostasis and potentially to the pathogenesis of neurodegenerative diseases by generating abnormal calcium ion release from the endoplasmic reticulum, according to Bruno et al. (Neurobiol Aging 33(5):1001 e1-6, 2012) and Stutzmann et al. (J Neurosci 24(2):508-13, 2004). Since ryanodine receptors share functional and structural similarities with potassium channels, as reported by Lanner et al. (Cold Spring Harb Perspect Biol 2(11):a003996, 2010), and small molecules with anti-oxidant properties, such as resveratrol (3,5,4'-trihydroxy-trans-stilbene), directly control the activity of potassium channels, according to Wang et al. (J Biomed Sci 23(1):47, 2016), McCalley et al. (Molecules 19(6):7327-40, 2014), Novakovic et al. (Mol Hum Reprod 21(6):545-51, 2015), Li et al. (Cardiovasc Res 45(4):1035-45, 2000), Gopalakrishnan et al. (Br J Pharmacol 129(7):1323-32, 2000), and Hambrock et al. (J Biol Chem 282(5):3347-56, 2007), we hypothesized that trans-resveratrol can modulate intracellular calcium signaling through direct binding and functional regulation of ryanodine receptors. The goal of our study was to identify and measure the control of ryanodine receptor activity by trans-resveratrol. Mechanisms of calcium signaling mediated by the direct interaction between trans-resveratrol and ryanodine receptors were identified and measured with single-channel electrophysiology. Addition of trans-resveratrol to the cytoplasmic face of the ryanodine receptor increased single-channel activity at physiological and elevated pathophysiological cytoplasmic calcium ion concentrations. The open probability of the channel increases after interacting with the small molecule in a dose-dependent manner, but remains also dependent on the concentration of its physiological ligand, cytoplasmic-free calcium ions. This study provides the first evidence of a direct functional interaction between trans-resveratrol and ryanodine receptors. Such functional control of ryanodine receptors by trans-resveratrol as a novel mechanism of action could provide additional rationales for the development of novel therapeutic strategies to treat and prevent neurodegenerative diseases.
Collapse
|
131
|
Chirasani VR, Xu L, Addis HG, Pasek DA, Dokholyan NV, Meissner G, Yamaguchi N. A central core disease mutation in the Ca 2+-binding site of skeletal muscle ryanodine receptor impairs single-channel regulation. Am J Physiol Cell Physiol 2019; 317:C358-C365. [PMID: 31166712 PMCID: PMC6732417 DOI: 10.1152/ajpcell.00052.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022]
Abstract
Cryoelectron microscopy and mutational analyses have shown that type 1 ryanodine receptor (RyR1) amino acid residues RyR1-E3893, -E3967, and -T5001 are critical for Ca2+-mediated activation of skeletal muscle Ca2+ release channel. De novo missense mutation RyR1-Q3970K in the secondary binding sphere of Ca2+ was reported in association with central core disease (CCD) in a 2-yr-old boy. Here, we characterized recombinant RyR1-Q3970K mutant by cellular Ca2+ release measurements, single-channel recordings, and computational methods. Caffeine-induced Ca2+ release studies indicated that RyR1-Q3970K formed caffeine-sensitive, Ca2+-conducting channel in HEK293 cells. However, in single-channel recordings, RyR1-Q3970K displayed low Ca2+-dependent channel activity and greatly reduced activation by caffeine or ATP. A RyR1-Q3970E mutant corresponds to missense mutation RyR2-Q3925E associated with arrhythmogenic syndrome in cardiac muscle. RyR1-Q3970E also formed caffeine-induced Ca2+ release in HEK293 cells and exhibited low activity in the presence of the activating ligand Ca2+ but, in contrast to RyR1-Q3970K, was activated by ATP and caffeine in single-channel recordings. Computational analyses suggested distinct structural rearrangements in the secondary binding sphere of Ca2+ of the two mutants, whereas the interaction of Ca2+ with directly interacting RyR1 amino acid residues Glu3893, Glu3967, and Thr5001 was only minimally affected. We conclude that RyR1-Q3970 has a critical role in Ca2+-dependent activation of RyR1 and that a missense RyR1-Q3970K mutant may give rise to myopathy in skeletal muscle.
Collapse
Affiliation(s)
- Venkat R Chirasani
- Departments of Pharmacology and Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Le Xu
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Hannah G Addis
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina
| | - Daniel A Pasek
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Nikolay V Dokholyan
- Departments of Pharmacology and Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Gerhard Meissner
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina
| | - Naohiro Yamaguchi
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina and Clemson University, Charleston, South Carolina
| |
Collapse
|
132
|
Wang T, Zhang L, Shi C, Wei R, Yin C. Interaction of the Homer1 EVH1 domain and skeletal muscle ryanodine receptor. Biochem Biophys Res Commun 2019; 514:720-725. [PMID: 31078268 DOI: 10.1016/j.bbrc.2019.04.199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
Abstract
The skeletal muscle ryanodine receptor (RyR1) proteins are intracellular calcium (Ca2+) release channels on the membrane of the sarcoplasmic reticulum (SR) and required for skeletal muscle excitation-contraction coupling. Homer (Vesl) is a family of scaffolding proteins that modulate target proteins including RyRs (ryanodine receptors), mGluRs (group 1 metabotropic glutamate receptors) and IP3Rs (inositol-1,4,5-trisphosphate receptors) through a conserved EVH1 (Ena/VASP homology 1) domain. Here, we examined the interaction between Homer1 EVH1 domain and RyR1 by co-immunoprecipitation, continuous sucrose density-gradient centrifugation, and bio-layer interferometry binding assay at different Ca2+ concentrations. Our results show that there exists a high-affinity binding between the Homer1 EVH1 domain and RyR1, especially at 1 mM of Ca2+. Based on our data and the known structures of Homer1 EVH1 domain and RyR1, we found two consensus proline-rich sequences in the structure of RyR1, PPHHF and FLPPP, and proposed two corresponding binding models to show mechanisms of recognition different from those used by other proline-rich motifs. The side proline residues of two proline-rich motifs from RyR1 are away from the hydrophobic surface of Homer1 EVH1, rather than buried in this hydrophobic surface. Our results provide evidence that Homer1 regulates RyR1 by direct interaction.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Biophysics, The Health Science Center, Peking University, Beijing, 100191, China.
| | - Lei Zhang
- Department of Biophysics, The Health Science Center, Peking University, Beijing, 100191, China; Electron Microscopy Analysis Laboratory, The Health Science Center, Peking University, Beijing, 100191, China.
| | - Chao Shi
- Department of Biophysics, The Health Science Center, Peking University, Beijing, 100191, China.
| | - Risheng Wei
- Department of Biophysics, The Health Science Center, Peking University, Beijing, 100191, China.
| | - Changcheng Yin
- Department of Biophysics, The Health Science Center, Peking University, Beijing, 100191, China; Electron Microscopy Analysis Laboratory, The Health Science Center, Peking University, Beijing, 100191, China; Center for Protein Science, Peking University, Beijing, 100871, China.
| |
Collapse
|
133
|
Three weeks of sprint interval training improved high-intensity cycling performance and limited ryanodine receptor modifications in recreationally active human subjects. Eur J Appl Physiol 2019; 119:1951-1958. [DOI: 10.1007/s00421-019-04183-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/22/2019] [Indexed: 01/04/2023]
|
134
|
Haji-Ghassemi O, Yuchi Z, Van Petegem F. The Cardiac Ryanodine Receptor Phosphorylation Hotspot Embraces PKA in a Phosphorylation-Dependent Manner. Mol Cell 2019; 75:39-52.e4. [PMID: 31078384 DOI: 10.1016/j.molcel.2019.04.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/05/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
Ryanodine receptors (RyRs) are intracellular Ca2+ release channels controlling essential cellular functions. RyRs are targeted by cyclic AMP (cAMP)-dependent protein kinase A (PKA), a controversial regulation implicated in disorders ranging from heart failure to Alzheimer's. Using crystal structures, we show that the phosphorylation hotspot domain of RyR2 embraces the PKA catalytic subunit, with an extensive interface not seen in PKA complexes with peptides. We trapped an intermediary open-form PKA bound to the RyR2 domain and an ATP analog, showing that PKA can engage substrates in an open form. Phosphomimetics or prior phosphorylation at nearby sites in RyR2 either enhance or reduce the activity of PKA. Finally, we show that a phosphomimetic at S2813, a well-known target site for calmodulin-dependent kinase II, induces the formation of an alpha helix in the phosphorylation domain, resulting in increased interactions and PKA activity. This shows that the different phosphorylation sites in RyR2 are not independent.
Collapse
Affiliation(s)
- Omid Haji-Ghassemi
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Zhiguang Yuchi
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
135
|
De Loof A, Schoofs L. Flip-Flopping Retinal in Microbial Rhodopsins as a Template for a Farnesyl/Prenyl Flip-Flop Model in Eukaryote GPCRs. Front Neurosci 2019; 13:465. [PMID: 31133794 PMCID: PMC6515946 DOI: 10.3389/fnins.2019.00465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Thirty years after the first description and modeling of G protein coupled receptors (GPCRs), information about their mode of action is still limited. One of the questions that is hard to answer is: how do the allosteric changes in the GPCR induced by, e.g., ligand binding in the end activate a G protein-dependent intracellular pathway (e.g., via the cAMP or the phosphatidylinositol signal pathways). Another question relates to the role of prenylation of G proteins. Today's "consensus model" states that protein prenylation is required for the assembly of GPCR-G protein complexes. Although it is well-known that protein prenylation is the covalent addition of a farnesyl- or geranylgeranyl moiety to the C terminus of specific proteins, e.g., α or γ G protein, the reason for this strong covalent binding remains enigmatic. The arguments for a fundamental role for prenylation of G proteins other than just being a hydrophobic linker, are gradually accumulating. We uncovered a dilemma that at first glance may be considered physiologically irrelevant, however, it may cause a true change in paradigm. The consensus model suggests that the only functional role of prenylation is to link the G protein to the receptor. Does the isoprenoid nature of the prenyl group and its exact site of attachment somehow matter? Or, are there valid arguments favoring the alternative possibility that a key role of the G protein is to guide the covalently attached prenyl group to - and it hold it in - a very specific location in between specific helices of the receptor? Our model says that the farnesyl/prenyl group - aided by its covalent attachment to a G protein -might function in GPCRs as a horseshoe-shaped flexible (and perhaps flip-flopping) hydrophobic valve for restricting (though not fully inhibiting) the untimely passage of Ca2+, like retinal does for the passage of H+ in microbial rhodopsins that are ancestral to many GPCRs.
Collapse
Affiliation(s)
- Arnold De Loof
- Functional Genomics and Proteomics Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
136
|
RYR1 Sequence Variants in Myopathies: Expression and Functional Studies in Two Families. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7638946. [PMID: 31165076 PMCID: PMC6500691 DOI: 10.1155/2019/7638946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/19/2019] [Indexed: 12/26/2022]
Abstract
The skeletal muscle ryanodine receptor (RyR1), i.e., the Ca2+ channel of the sarco/endoplasmic reticulum (S/ER), and the voltage-dependent calcium channel Cav1.1 are the principal channels involved in excitation-contraction coupling in skeletal muscle. RYR1 gene variants are linked to distinct skeletal muscle disorders, including malignant hyperthermia susceptibility and central core disease (CCD), mainly with autosomal dominant inheritance, and autosomal recessive myopathies with a broad phenotypic and histopathological spectrum. The age at onset of RYR1-related myopathies varies from infancy to adulthood. We report the identification of four RYR1 variants in two Italian families: one with myopathy and variants c.4003C>T (p.R1335C) and c.7035C>A (p.S2345R), and another with CCD and variants c.9293G>T (p.S3098I) and c.14771_14772insTAGACAGGGTGTTGCTCTGTTGCCCTTCTT (p.F4924_V4925insRQGVALLPFF). We demonstrate that, in patient-specific lymphoblastoid cells, the c.4003C>T (p.R1335C) variant is not expressed and the in-frame 30-nucleotide insertion variant is expressed at a low level. Moreover, Ca2+ release in response to the RyR1 agonist 4-chloro-m-cresol and to thapsigargin showed that the c.7035C>A (p.S2345R) variant causes depletion of S/ER Ca2+ stores and that the compound heterozygosity for variant c.9293G>T (p.S3098I) and the 30-nucleotide insertion increases RyR1-dependent Ca2+ release without affecting ER Ca2+ stores. In conclusion, we detected and functionally characterized disease-causing variants of the RyR1 channel in patient-specific lymphoblastoid cells. This paper is dedicated to the memory and contribution of Luigi Del Vecchio.
Collapse
|
137
|
Taylor CW, Machaca K. IP3 receptors and store-operated Ca2+ entry: a license to fill. Curr Opin Cell Biol 2019; 57:1-7. [DOI: 10.1016/j.ceb.2018.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/05/2018] [Indexed: 10/28/2022]
|
138
|
Prole DL, Taylor CW. Structure and Function of IP 3 Receptors. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035063. [PMID: 30745293 DOI: 10.1101/cshperspect.a035063] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs), by releasing Ca2+ from the endoplasmic reticulum (ER) of animal cells, allow Ca2+ to be redistributed from the ER to the cytosol or other organelles, and they initiate store-operated Ca2+ entry (SOCE). For all three IP3R subtypes, binding of IP3 primes them to bind Ca2+, which then triggers channel opening. We are now close to understanding the structural basis of IP3R activation. Ca2+-induced Ca2+ release regulated by IP3 allows IP3Rs to regeneratively propagate Ca2+ signals. The smallest of these regenerative events is a Ca2+ puff, which arises from the nearly simultaneous opening of a small cluster of IP3Rs. Ca2+ puffs are the basic building blocks for all IP3-evoked Ca2+ signals, but only some IP3 clusters, namely those parked alongside the ER-plasma membrane junctions where SOCE occurs, are licensed to respond. The location of these licensed IP3Rs may allow them to selectively regulate SOCE.
Collapse
Affiliation(s)
- David L Prole
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| |
Collapse
|
139
|
Ríos E, Gillespie D, Franzini-Armstrong C. The binding interactions that maintain excitation-contraction coupling junctions in skeletal muscle. J Gen Physiol 2019; 151:593-605. [PMID: 30728215 PMCID: PMC6445584 DOI: 10.1085/jgp.201812268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/02/2019] [Indexed: 12/03/2022] Open
Abstract
Calcium for contraction of skeletal muscles is released via tetrameric ryanodine receptor (RYR1) channels of the sarcoplasmic reticulum (SR), which are assembled in ordered arrays called couplons at junctions where the SR abuts T tubules or plasmalemma. Voltage-gated Ca2+ (CaV1.1) channels, found in tubules or plasmalemma, form symmetric complexes called CaV tetrads that associate with and activate underlying RYR tetramers during membrane depolarization by conveying a conformational change. Intriguingly, CaV tetrads regularly skip every other RYR tetramer within the array; therefore, the RYRs underlying tetrads (named V), but not the voltage sensor-lacking (C) RYRs, should be activated by depolarization. Here we hypothesize that the checkerboard association is maintained solely by reversible binary interactions between CaVs and RYRs and test this hypothesis using a quantitative model of the energies that govern CaV1.1-RYR1 binding, which are assumed to depend on number and location of bound CaVs. A Monte Carlo simulation generates large statistical samples and distributions of state variables that can be compared with quantitative features in freeze-fracture images of couplons from various sources. This analysis reveals two necessary model features: (1) the energy of a tetramer must have wells at low and high occupation by CaVs, so that CaVs positively cooperate in binding RYR (an allosteric effect), and (2) a large energy penalty results when two CaVs bind simultaneously to adjacent RYR protomers in adjacent tetramers (a steric clash). Under the hypothesis, V and C channels will eventually reverse roles. Role reversal justifies the presence of sensor-lacking C channels, as a structural and functional reserve for control of muscle contraction.
Collapse
Affiliation(s)
- Eduardo Ríos
- Section of Cellular Signaling, Department of Physiology and Biophysics, Rush University, Chicago, IL
| | - Dirk Gillespie
- Section of Cellular Signaling, Department of Physiology and Biophysics, Rush University, Chicago, IL
| | | |
Collapse
|
140
|
Gutiérrez-Moreno R, Mota-Sanchez D, Blanco CA, Whalon ME, Terán-Santofimio H, Rodriguez-Maciel JC, DiFonzo C. Field-Evolved Resistance of the Fall Armyworm (Lepidoptera: Noctuidae) to Synthetic Insecticides in Puerto Rico and Mexico. JOURNAL OF ECONOMIC ENTOMOLOGY 2019; 112:792-802. [PMID: 30535077 DOI: 10.1093/jee/toy372] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Indexed: 05/18/2023]
Abstract
The fall armyworm, Spodoptera frugiperda (J. E. Smith), is one of the main pests of corn in many areas of the American continent. The reliance on pesticides to control fall armyworm has led to the development of insecticide resistance in many regions. We determined the resistance levels of fall armyworm to insecticides of different modes of action in fall armyworm populations from Puerto Rico and several Mexican states with different insecticide use patterns. Mexican populations that expressed higher resistance ratios (RR50) were: Sonora (20-fold to chlorpyriphos), Oaxaca (19-fold to permethrin), and Sinaloa (10-fold to flubendamide). The Puerto Rico population exhibited a remarkable field-evolved resistance to many pesticides. The RR50 to the insecticides tested were: flubendiamide (500-fold), chlorantraniliprole (160-fold), methomyl (223-fold), thiodicarb (124-fold), permethrin (48-fold), chlorpyriphos (47-fold), zeta-cypermethrin (35-fold), deltamethrin (25-fold), triflumuron (20-fold), spinetoram (14-fold). Spinosad (eightfold), emamectin benzoate and abamectin (sevenfold) displayed lower resistance ratio. However, these compounds are still effective to manage fall armyworm resistance in Puerto Rico. Fall armyworm populations from Mexico show different levels of susceptibility, which may reflect the heterogeneity of the pest control patterns in this country. The status of insecticide resistance in the fall armyworm from Puerto Rico indicates a challenging situation for the control of this pest with these insecticides in the close future. Lessons learned from this research might be applied in regions with recent invasions of fall armyworm in Africa.
Collapse
Affiliation(s)
- Rebeca Gutiérrez-Moreno
- Department of Entomology, Michigan State University, Food Safety and Toxicology Building, East Lansing, MI
| | - David Mota-Sanchez
- Department of Entomology, Michigan State University, Food Safety and Toxicology Building, East Lansing, MI
| | - Carlos A Blanco
- Department of Biology, University of New Mexico, Albuquerque, Mexico
| | - Mark E Whalon
- Department of Entomology, Michigan State University, Food Safety and Toxicology Building, East Lansing, MI
| | | | | | - Christina DiFonzo
- Department of Entomology, Michigan State University, Food Safety and Toxicology Building, East Lansing, MI
| |
Collapse
|
141
|
Koenig X, Choi RH, Schicker K, Singh DP, Hilber K, Launikonis BS. Mechanistic insights into store-operated Ca 2+ entry during excitation-contraction coupling in skeletal muscle. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1239-1248. [PMID: 30825472 DOI: 10.1016/j.bbamcr.2019.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 01/11/2023]
Abstract
Skeletal muscle fibres support store-operated Ca2+-entry (SOCE) across the t-tubular membrane upon exhaustive depletion of Ca2+ from the sarcoplasmic reticulum (SR). Recently we demonstrated the presence of a novel mode of SOCE activated under conditions of maintained [Ca2+]SR. This phasic SOCE manifested in a fast and transient manner in synchrony with excitation contraction (EC)-coupling mediated SR Ca2+-release (Communications Biology 1:31, doi: https://doi.org/10.1038/s42003-018-0033-7). Stromal interaction molecule 1 (STIM1) and calcium release-activated calcium channel 1 (ORAI1), positioned at the SR and t-system membranes, respectively, are the considered molecular correlate of SOCE. The evidence suggests that at the triads, where the terminal cisternae of the SR sandwich a t-tubule, STIM1 and ORAI1 proteins pre-position to allow for enhanced SOCE transduction. Here we show that phasic SOCE is not only shaped by global [Ca2+]SR but provide evidence for a local activation within nanodomains at the terminal cisternae of the SR. This feature may allow SOCE to modulate [Ca2+]SR during EC coupling. We define SOCE to occur on the same timescale as EC coupling and determine the temporal coherence of SOCE activation to SR Ca2+ release. We derive a delay of 0.3 ms reflecting diffusive Ca2+-equilibration at the luminal ryanodine receptor 1 (RyR1) channel mouth upon SR Ca2+-release. Numerical simulations of Ca2+-calsequestrin binding estimates a characteristic diffusion length and confines an upper limit for the spatial distance between STIM1 and RyR1. Experimental evidence for a 4- fold change in t-system Ca2+-permeability upon prolonged electrical stimulation in conjunction with numerical simulations of Ca2+-STIM1 binding suggests a Ca2+ dissociation constant of STIM1 below 0.35 mM. Our results show that phasic SOCE is intimately linked with RyR opening and closing, with only μs delays, because [Ca2+] in the terminal cisternae is just above the threshold for Ca2+ dissociation from STIM1 under physiological resting conditions. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Xaver Koenig
- Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Wien, Austria.
| | - Rocky H Choi
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Klaus Schicker
- Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Wien, Austria
| | - Daniel P Singh
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Karlheinz Hilber
- Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Wien, Austria
| | - Bradley S Launikonis
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
142
|
Shekar S, Chien CC, Hartel A, Ong P, Clarke OB, Marks A, Drndic M, Shepard KL. Wavelet Denoising of High-Bandwidth Nanopore and Ion-Channel Signals. NANO LETTERS 2019; 19:1090-1097. [PMID: 30601669 PMCID: PMC6904930 DOI: 10.1021/acs.nanolett.8b04388] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Recent work has pushed the noise-limited bandwidths of solid-state nanopore conductance recordings to more than 5 MHz and of ion channel conductance recordings to more than 500 kHz through the use of integrated complementary metal-oxide-semiconductor (CMOS) integrated circuits. Despite the spectral spread of the pulse-like signals that characterize these recordings when a sinusoidal basis is employed, Bessel filters are commonly used to denoise these signals to acceptable signal-to-noise ratios (SNRs) at the cost of losing many of the faster temporal features. Here, we report improvements to the SNR that can be achieved using wavelet denoising instead of Bessel filtering. When combined with state-of-the-art high-bandwidth CMOS recording instrumentation, we can reduce baseline noise levels by over a factor of 4 compared to a 2.5 MHz Bessel filter while retaining transient properties in the signal comparable to this filter bandwidth. Similarly, for ion-channel recordings, we achieve a temporal response better than a 100 kHz Bessel filter with a noise level comparable to that achievable with a 25 kHz Bessel filter. Improvements in SNR can be used to achieve robust statistical analyses of these recordings, which may provide important insights into nanopore translocation dynamics and mechanisms of ion-channel function.
Collapse
Affiliation(s)
| | - Chen-Chi Chien
- Department of Physics and Astronomy , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | | | | | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics , Columbia University , New York , New York 10032 , United States
| | - Andrew Marks
- Department of Physiology and Cellular Biophysics , Columbia University , New York , New York 10032 , United States
| | - Marija Drndic
- Department of Physics and Astronomy , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | | |
Collapse
|
143
|
Ca 2+-dependent calmodulin binding to cardiac ryanodine receptor (RyR2) calmodulin-binding domains. Biochem J 2019; 476:193-209. [PMID: 30530841 PMCID: PMC6340113 DOI: 10.1042/bcj20180545] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/05/2018] [Accepted: 12/10/2018] [Indexed: 12/17/2022]
Abstract
The Ca2+ sensor calmodulin (CaM) regulates cardiac ryanodine receptor (RyR2)-mediated Ca2+ release from the sarcoplasmic reticulum. CaM inhibits RyR2 in a Ca2+-dependent manner and aberrant CaM-dependent inhibition results in life-threatening cardiac arrhythmias. However, the molecular details of the CaM–RyR2 interaction remain unclear. Four CaM-binding domains (CaMBD1a, -1b, -2, and -3) in RyR2 have been proposed. Here, we investigated the Ca2+-dependent interactions between CaM and these CaMBDs by monitoring changes in the fluorescence anisotropy of carboxytetramethylrhodamine (TAMRA)-labeled CaMBD peptides during titration with CaM at a wide range of Ca2+ concentrations. We showed that CaM bound to all four CaMBDs with affinities that increased with Ca2+ concentration. CaM bound to CaMBD2 and -3 with high affinities across all Ca2+ concentrations tested, but bound to CaMBD1a and -1b only at Ca2+ concentrations above 0.2 µM. Binding experiments using individual CaM domains revealed that the CaM C-domain preferentially bound to CaMBD2, and the N-domain to CaMBD3. Moreover, the Ca2+ affinity of the CaM C-domain in complex with CaMBD2 or -3 was so high that these complexes are essentially Ca2+ saturated under resting Ca2+ conditions. Conversely, the N-domain senses Ca2+ exactly in the transition from resting to activating Ca2+ when complexed to either CaMBD2 or -3. Altogether, our results support a binding model where the CaM C-domain is anchored to RyR2 CaMBD2 and saturated with Ca2+ during Ca2+ oscillations, while the CaM N-domain functions as a dynamic Ca2+ sensor that can bridge noncontiguous regions of RyR2 or clamp down onto CaMBD2.
Collapse
|
144
|
Ivarsson N, Mattsson CM, Cheng AJ, Bruton JD, Ekblom B, Lanner JT, Westerblad H. SR Ca 2+ leak in skeletal muscle fibers acts as an intracellular signal to increase fatigue resistance. J Gen Physiol 2019; 151:567-577. [PMID: 30635368 PMCID: PMC6445590 DOI: 10.1085/jgp.201812152] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/16/2018] [Indexed: 01/14/2023] Open
Abstract
Skeletal muscle oxidative capacity and fatigue resistance can be improved with endurance training, but the mechanism is not fully understood. Ivarsson et al. find that the signaling pathway that increases fatigue resistance in muscle is triggered by a mild Ca2+ leak from the sarcoplasmic reticulum. Effective practices to improve skeletal muscle fatigue resistance are crucial for athletes as well as patients with dysfunctional muscles. To this end, it is important to identify the cellular signaling pathway that triggers mitochondrial biogenesis and thereby increases oxidative capacity and fatigue resistance in skeletal muscle fibers. Here, we test the hypothesis that the stress induced in skeletal muscle fibers by endurance exercise causes a reduction in the association of FK506-binding protein 12 (FKBP12) with ryanodine receptor 1 (RYR1). This will result in a mild Ca2+ leak from the sarcoplasmic reticulum (SR), which could trigger mitochondrial biogenesis and improved fatigue resistance. After giving mice access to an in-cage running wheel for three weeks, we observed decreased FKBP12 association to RYR1, increased baseline [Ca2+]i, and signaling associated with greater mitochondrial biogenesis in muscle, including PGC1α1. After six weeks of voluntary running, FKBP12 association is normalized, baseline [Ca2+]i returned to values below that of nonrunning controls, and signaling for increased mitochondrial biogenesis was no longer present. The adaptations toward improved endurance exercise performance that were observed with training could be mimicked by pharmacological agents that destabilize RYR1 and thereby induce a modest Ca2+ leak. We conclude that a mild RYR1 SR Ca2+ leak is a key trigger for the signaling pathway that increases muscle fatigue resistance.
Collapse
Affiliation(s)
- Niklas Ivarsson
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Stockholm, Sweden
| | - C Mikael Mattsson
- Åstrand Laboratory of Work Physiology, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Arthur J Cheng
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Stockholm, Sweden
| | - Joseph D Bruton
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Stockholm, Sweden
| | - Björn Ekblom
- Åstrand Laboratory of Work Physiology, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
145
|
Steele TWE, Samsó M. The FKBP12 subunit modifies the long-range allosterism of the ryanodine receptor. J Struct Biol 2019; 205:180-188. [PMID: 30641143 DOI: 10.1016/j.jsb.2018.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023]
Abstract
Ryanodine receptors (RyRs) are large conductance intracellular channels controlling intracellular calcium homeostasis in myocytes, neurons, and other cell types. Loss of RyR's constitutive cytoplasmic partner FKBP results in channel sensitization, dominant subconductance states, and increased cytoplasmic Ca2+. FKBP12 binds to RyR1's cytoplasmic assembly 130 Å away from the ion gate at four equivalent sites in the RyR1 tetramer. To understand how FKBP12 binding alters RyR1's channel properties, we studied the 3D structure of RyR1 alone in the closed conformation in the context of the open and closed conformations of FKBP12-bound RyR1. We analyzed the metrics of conformational changes of existing structures, the structure of the ion gate, and carried out multivariate statistical analysis of thousands of individual cryoEM RyR1 particles. We find that under closed state conditions, in the presence of FKBP12, the cytoplasmic domain of RyR1 adopts an upward conformation, whereas absence of FKBP12 results in a relaxed conformation, while the ion gate remains closed. The relaxed conformation is intermediate between the RyR1-FKBP12 complex closed (upward) and open (downward) conformations. The closed-relaxed conformation of RyR1 appears to be consistent with a lower energy barrier separating the closed and open states of RyR1-FKBP12, and suggests that FKBP12 plays an important role by restricting conformations within RyR1's conformational landscape.
Collapse
Affiliation(s)
- Tyler W E Steele
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Montserrat Samsó
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|
146
|
Feng R, Zhou X, Zhang W, Pu T, Sun Y, Yang R, Wang D, Zhang X, Gao Y, Cai Z, Liang Y, Yu Q, Wu Y, Lei X, Liang Z, Jones O, Wang L, Xu M, Sun Y, Isaacs WB, Ma J, Xu X. Dynamics expression of DmFKBP12/Calstabin during embryonic early development of Drosophila melanogaster. Cell Biosci 2019; 9:8. [PMID: 30637096 PMCID: PMC6325743 DOI: 10.1186/s13578-019-0270-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022] Open
Abstract
Background Calcium signaling are conserved from invertebrates to vertebrates and plays critical roles in many molecular mechanisms of embryogenesis and postnatal development. As a critical component of the signaling pathway, the RyR medicated calcium-induced calcium release signaling system, has been well studied along with their regulator FK506-binding protein 12 (FKBP12/Calstabin). Lack of FKBP12 is known to result in lethal cardiac dysfunction in mouse. However, precisely how FKBP12 is regulated and effects calcium signaling in Drosophila melanogaster remains largely unknown. Results In this study, we identified both temporal and localization changes in expression of DmFKBP12, a translational and transcriptional regulator of Drosophila RyR (DmRyR) and FKBP12, through embryonic development. DmFKBP12 is first expressed at the syncytial blastoderm stage and undergoes increased expression during the cellular blastoderm and early gastrulation stages. At late gastrulation, DmFKBP12 expression begins to decline until it reaches homeostasis, which it then maintains throughout the rest of development. Throughout these described changes in expression, DmFKBP12 mRNA remain stable, which indicates that protein dynamics are attributed to regulation at the mRNA to protein translation level. In addition to temporal changes in expression, dynamic expression profiles during Drosophila development also revealed DmFKBP12 localization. Although DmFKBP12 is distributed evenly between the anterior to posterior poles of the blastoderm egg, the protein is expressed more strongly in the cortex of the early Drosophila gastrula with the highest concentration found in the basement membrane of the cellular blastoderm. Fertilized egg, through the profile as under-membrane cortex distribution concentering onto basement at cellular blastoderm, to the profile as three-gem layer localization in primitive neuronal and digestion architecture of early Drosophila gastrula. By late gastrulation, DmFKBP12 is no longer identified in the yolk or lumen of duct structures and has relocated to the future brain (suboesophageal and supraesophageal ganglions), ventral nervous system, and muscular system. Throughout these changes in distribution, in situ DmFKBP12 mRNA monitoring detected equal distribution of DmFKBP12 mRNA, once again indicating that regulation of DmFKBP12 occurs at the translational level in Drosophila development. Conclusion As a critical regulator of the DmRyR-FKBP complex, DmFKBP12 expression in Drosophila fluctuates temporally and geographically with the formation of organ systems. These finding indicate that DmFKBP12 and RyR associated calcium signaling plays an essential role in the successful development of Drosophila melanogaster. Further study on the differences between mammalian RyR-FKBP12 and Drosophila DmRyR-FKBP12 can be exploited to develop safe pesticides. Electronic supplementary material The online version of this article (10.1186/s13578-019-0270-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Feng
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Xin Zhou
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China.,2Ohio State University School of Medicine, Columbus, OH 43210 USA
| | - Wei Zhang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Tao Pu
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Yuting Sun
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Rong Yang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Dan Wang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Xiaofei Zhang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Yingfeng Gao
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Zhenlu Cai
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Yu Liang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Qiuxia Yu
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Yajun Wu
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Xinjuan Lei
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Zhijia Liang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| | - Odell Jones
- 4University of Pennsylvania ULAR, Philadelphia, PA 19144 USA
| | - Liyang Wang
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China.,Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215 USA
| | - Mengmeng Xu
- 5Medical-Scientist Training Program, Department of Pharmacology, Duke University Medical Center, Durham, NC 27710 USA
| | - Yanping Sun
- 6College of Pharmacy, Xi'an Medical University, Xi'an, 710062 China
| | | | - Jianjie Ma
- 2Ohio State University School of Medicine, Columbus, OH 43210 USA
| | - Xuehong Xu
- 1National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China/CGDB, Shaanxi Normal University College of Life Sciences, Xi'an, 710062 China
| |
Collapse
|
147
|
Chen J, Xue L, Wei R, Liu S, Yin CC. The insecticide chlorantraniliprole is a weak activator of mammalian skeletal ryanodine receptor/Ca2+ release channel. Biochem Biophys Res Commun 2019; 508:633-639. [DOI: 10.1016/j.bbrc.2018.11.180] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022]
|
148
|
Rossi AM, Taylor CW. IP3 receptors – lessons from analyses ex cellula. J Cell Sci 2018; 132:132/4/jcs222463. [DOI: 10.1242/jcs.222463] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
ABSTRACT
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are widely expressed intracellular channels that release Ca2+ from the endoplasmic reticulum (ER). We review how studies of IP3Rs removed from their intracellular environment (‘ex cellula’), alongside similar analyses of ryanodine receptors, have contributed to understanding IP3R behaviour. Analyses of permeabilized cells have demonstrated that the ER is the major intracellular Ca2+ store, and that IP3 stimulates Ca2+ release from this store. Radioligand binding confirmed that the 4,5-phosphates of IP3 are essential for activating IP3Rs, and facilitated IP3R purification and cloning, which paved the way for structural analyses. Reconstitution of IP3Rs into lipid bilayers and patch-clamp recording from the nuclear envelope have established that IP3Rs have a large conductance and select weakly between Ca2+ and other cations. Structural analyses are now revealing how IP3 binding to the N-terminus of the tetrameric IP3R opens the pore ∼7 nm away from the IP3-binding core (IBC). Communication between the IBC and pore passes through a nexus of interleaved domains contributed by structures associated with the pore and cytosolic domains, which together contribute to a Ca2+-binding site. These structural analyses provide evidence to support the suggestion that IP3 gates IP3Rs by first stimulating Ca2+ binding, which leads to pore opening and Ca2+ release.
Collapse
Affiliation(s)
- Ana M. Rossi
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Colin W. Taylor
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| |
Collapse
|
149
|
Dulhunty AF, Beard NA, Casarotto MG. Recent advances in understanding the ryanodine receptor calcium release channels and their role in calcium signalling. F1000Res 2018; 7. [PMID: 30542613 PMCID: PMC6259491 DOI: 10.12688/f1000research.16434.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2018] [Indexed: 12/30/2022] Open
Abstract
The ryanodine receptor calcium release channel is central to cytoplasmic Ca
2+ signalling in skeletal muscle, the heart, and many other tissues, including the central nervous system, lymphocytes, stomach, kidney, adrenal glands, ovaries, testes, thymus, and lungs. The ion channel protein is massive (more than 2.2 MDa) and has a structure that has defied detailed determination until recent developments in cryo-electron microscopy revealed much of its structure at near-atomic resolution. The availability of this high-resolution structure has provided the most significant advances in understanding the function of the ion channel in the past 30 years. We can now visualise the molecular environment of individual amino acid residues that form binding sites for essential modulators of ion channel function and determine its role in Ca
2+ signalling. Importantly, the structure has revealed the structural environment of the many deletions and point mutations that disrupt Ca
2+ signalling in skeletal and cardiac myopathies and neuropathies. The implications are of vital importance to our understanding of the molecular basis of the ion channel’s function and for the design of therapies to counteract the effects of ryanodine receptor-associated disorders.
Collapse
Affiliation(s)
- Angela F Dulhunty
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, 131 Garran Road, The Australian National University, Acton, ACT, 2601, Australia
| | - Nicole A Beard
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Bruce, ACT, 2617, Australia
| | - Marco G Casarotto
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, 131 Garran Road, The Australian National University, Acton, ACT, 2601, Australia
| |
Collapse
|
150
|
Acimovic I, Refaat MM, Moreau A, Salykin A, Reiken S, Sleiman Y, Souidi M, Přibyl J, Kajava AV, Richard S, Lu JT, Chevalier P, Skládal P, Dvořak P, Rotrekl V, Marks AR, Scheinman MM, Lacampagne A, Meli AC. Post-Translational Modifications and Diastolic Calcium Leak Associated to the Novel RyR2-D3638A Mutation Lead to CPVT in Patient-Specific hiPSC-Derived Cardiomyocytes. J Clin Med 2018; 7:jcm7110423. [PMID: 30413023 PMCID: PMC6262462 DOI: 10.3390/jcm7110423] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 10/31/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Sarcoplasmic reticulum Ca2+ leak and post-translational modifications under stress have been implicated in catecholaminergic polymorphic ventricular tachycardia (CPVT), a highly lethal inherited arrhythmogenic disorder. Human induced pluripotent stem cells (hiPSCs) offer a unique opportunity for disease modeling. Objective: The aims were to obtain functional hiPSC-derived cardiomyocytes from a CPVT patient harboring a novel ryanodine receptor (RyR2) mutation and model the syndrome, drug responses and investigate the molecular mechanisms associated to the CPVT syndrome. Methods: Patient-specific cardiomyocytes were generated from a young athletic female diagnosed with CPVT. The contractile, intracellular Ca2+ handling and electrophysiological properties as well as the RyR2 macromolecular remodeling were studied. Results: Exercise stress electrocardiography revealed polymorphic ventricular tachycardia when treated with metoprolol and marked improvement with flecainide alone. We found abnormal stress-induced contractile and electrophysiological properties associated with sarcoplasmic reticulum Ca2+ leak in CPVT hiPSC-derived cardiomyocytes. We found inadequate response to metoprolol and a potent response of flecainide. Stabilizing RyR2 with a Rycal compound prevents those abnormalities specifically in CPVT hiPSC-derived cardiomyocytes. The RyR2-D3638A mutation is located in the conformational change inducing-central core domain and leads to RyR2 macromolecular remodeling including depletion of PP2A and Calstabin2. Conclusion: We identified a novel RyR2-D3638A mutation causing 3D conformational defects and aberrant biophysical properties associated to RyR2 macromolecular complex post-translational remodeling. The molecular remodeling is for the first time revealed using patient-specific hiPSC-derived cardiomyocytes which may explain the CPVT proband’s resistance. Our study promotes hiPSC-derived cardiomyocytes as a suitable model for disease modeling, testing new therapeutic compounds, personalized medicine and deciphering underlying molecular mechanisms.
Collapse
Affiliation(s)
- Ivana Acimovic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic.
| | - Marwan M Refaat
- Department of Internal Medicine, Cardiology Division/Cardiac Electrophysiology Section and Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center, Beirut 1107 2020, Lebanon.
| | - Adrien Moreau
- NeuroMyoGène Institute, University of Claude Bernard Lyon 1, 69100 Villeurbanne, France.
- PhyMedExp, INSERM, University of Montpellier, CNRS, 371 Avenue du Doyen G. Giraud, 34295 Montpellier CEDEX 5, France.
| | - Anton Salykin
- Department of Biology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic.
| | - Steve Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | - Yvonne Sleiman
- PhyMedExp, INSERM, University of Montpellier, CNRS, 371 Avenue du Doyen G. Giraud, 34295 Montpellier CEDEX 5, France.
| | - Monia Souidi
- PhyMedExp, INSERM, University of Montpellier, CNRS, 371 Avenue du Doyen G. Giraud, 34295 Montpellier CEDEX 5, France.
| | - Jan Přibyl
- CEITEC, Masaryk University, Brno 62500, Czech Republic.
| | - Andrey V Kajava
- CRBM, CNRS, University of Montpellier, 34293 Montpellier, France and University ITMO, St Petersburg 197101, Russia.
| | - Sylvain Richard
- PhyMedExp, INSERM, University of Montpellier, CNRS, 371 Avenue du Doyen G. Giraud, 34295 Montpellier CEDEX 5, France.
| | - Jonathan T Lu
- Department of Cardiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA.
| | - Philippe Chevalier
- NeuroMyoGène Institute, University of Claude Bernard Lyon 1, 69100 Villeurbanne, France.
| | - Petr Skládal
- CEITEC, Masaryk University, Brno 62500, Czech Republic.
| | - Petr Dvořak
- Department of Biology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic.
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic.
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | - Melvin M Scheinman
- San Francisco Medical Center, University of California, San Francisco, CA 94115, USA.
| | - Alain Lacampagne
- PhyMedExp, INSERM, University of Montpellier, CNRS, 371 Avenue du Doyen G. Giraud, 34295 Montpellier CEDEX 5, France.
| | - Albano C Meli
- PhyMedExp, INSERM, University of Montpellier, CNRS, 371 Avenue du Doyen G. Giraud, 34295 Montpellier CEDEX 5, France.
| |
Collapse
|