1
|
Euden J, Mason SA, Williams AJ. Functional characterization of the cardiac ryanodine receptor pore-forming region. PLoS One 2013; 8:e66542. [PMID: 23776685 PMCID: PMC3680380 DOI: 10.1371/journal.pone.0066542] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/07/2013] [Indexed: 01/16/2023] Open
Abstract
Ryanodine receptors are homotetrameric intracellular calcium release channels. The efficiency of these channels is underpinned by exceptional rates of cation translocation through the open channel and this is achieved at the expense of the high degree of selectivity characteristic of many other types of channel. Crystallization of prokaryotic potassium channels has provided insights into the structures and mechanisms responsible for ion selection and movement in these channels, however no equivalent structural detail is currently available for ryanodine receptors. Nevertheless both molecular modeling and cryo-electron microscopy have identified the probable pore-forming region (PFR) of the ryanodine receptor (RyR) and suggest that this region contains structural elements equivalent to those of the PFRs of potassium-selective channels. The aim of the current study was to establish if the isolated putative cardiac RyR (RyR2) PFR could form a functional ion channel. We have expressed and purified the RyR2 PFR and shown that function is retained following reconstitution into planar phospholipid bilayers. Our data provide the first direct experimental evidence to support the proposal that the conduction pathway of RyR2 is formed by structural elements equivalent to those of the potassium channel PFR.
Collapse
Affiliation(s)
- Joanne Euden
- Institute of Molecular and Experimental Medicine, Cardiff University, Cardiff, United Kingdom.
| | | | | |
Collapse
|
2
|
Venturi E, Matyjaszkiewicz A, Pitt SJ, Tsaneva-Atanasova K, Nishi M, Yamazaki D, Takeshima H, Sitsapesan R. TRIC-B channels display labile gating: evidence from the TRIC-A knockout mouse model. Pflugers Arch 2013; 465:1135-48. [PMID: 23467973 PMCID: PMC3732801 DOI: 10.1007/s00424-013-1251-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/24/2013] [Accepted: 02/11/2013] [Indexed: 11/25/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum (SR) and nuclear membranes contain two related cation channels named TRIC-A and TRIC-B. In many tissues, both subtypes are co-expressed, making it impossible to distinguish the distinct single-channel properties of each subtype. We therefore incorporated skeletal muscle SR vesicles derived from Tric-a-knockout mice into bilayers in order to characterise the biophysical properties of native TRIC-B without possible misclassification of the channels as TRIC-A, and without potential distortion of functional properties by detergent purification protocols. The native TRIC-B channels were ideally selective for cations. In symmetrical 210 mM K+, the maximum (full) open channel level (199 pS) was equivalent to that observed when wild-type SR vesicles were incorporated into bilayers. Analysis of TRIC-B gating revealed complex and variable behaviour. Four main sub-conductance levels were observed at approximately 80 % (161 pS), 60 % (123 pS), 46 % (93 pS), and 30 % (60 pS) of the full open state. Seventy-five percent of the channels were voltage sensitive with Po being markedly reduced at negative holding potentials. The frequent, rapid transitions between TRIC-B sub-conductance states prevented development of reliable gating models using conventional single-channel analysis. Instead, we used mean-variance plots to highlight key features of TRIC-B gating in a more accurate and visually useful manner. Our study provides the first biophysical characterisation of native TRIC-B channels and indicates that this channel would be suited to provide counter current in response to Ca2+ release from the SR. Further experiments are required to distinguish the distinct functional properties of TRIC-A and TRIC-B and understand their individual but complementary physiological roles.
Collapse
Affiliation(s)
- Elisa Venturi
- School of Physiology & Pharmacology, NSQI and Bristol Heart Institute, University of Bristol, Bristol, BS8 1TD UK
| | - Antoni Matyjaszkiewicz
- School of Physiology & Pharmacology, NSQI and Bristol Heart Institute, University of Bristol, Bristol, BS8 1TD UK
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1TR UK
- Bristol Centre for Complexity Sciences, University of Bristol, Bristol, BS8 1TR UK
| | - Samantha J. Pitt
- School of Medicine, University of St Andrews, St Andrews, KY16 9TF UK
| | | | - Miyuki Nishi
- Graduate School of Pharmaceutical Sciences and Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, 606-8501 Japan
| | - Daiju Yamazaki
- Graduate School of Pharmaceutical Sciences and Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, 606-8501 Japan
| | - Hiroshi Takeshima
- Graduate School of Pharmaceutical Sciences and Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, 606-8501 Japan
| | - Rebecca Sitsapesan
- School of Physiology & Pharmacology, NSQI and Bristol Heart Institute, University of Bristol, Bristol, BS8 1TD UK
| |
Collapse
|
3
|
Ramachandran S, Chakraborty A, Xu L, Mei Y, Samsó M, Dokholyan NV, Meissner G. Structural determinants of skeletal muscle ryanodine receptor gating. J Biol Chem 2013; 288:6154-65. [PMID: 23319589 DOI: 10.1074/jbc.m112.433789] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ryanodine receptor type 1 (RyR1) releases Ca(2+) from intracellular stores upon nerve impulse to trigger skeletal muscle contraction. Effector binding at the cytoplasmic domain tightly controls gating of the pore domain of RyR1 to release Ca(2+). However, the molecular mechanism that links effector binding to channel gating is unknown due to lack of structural data. Here, we used a combination of computational and electrophysiological methods and cryo-EM densities to generate structural models of the open and closed states of RyR1. Using our structural models, we identified an interface between the pore-lining helix (Tyr-4912-Glu-4948) and a linker helix (Val-4830-Val-4841) that lies parallel to the cytoplasmic membrane leaflet. To test the hypothesis that this interface controls RyR1 gating, we designed mutations in the linker helix to stabilize either the open (V4830W and T4840W) or closed (H4832W and G4834W) state and validated them using single channel experiments. To further confirm this interface, we designed mutations in the pore-lining helix to stabilize the closed state (Q4947N, Q4947T, and Q4947S), which we also validated using single channel experiments. The channel conductance and selectivity of the mutations that we designed in the linker and pore-lining helices were indistinguishable from those of WT RyR1, demonstrating our ability to modulate RyR1 gating without affecting ion permeation. Our integrated computational and experimental approach significantly advances the understanding of the structure and function of an unusually large ion channel.
Collapse
Affiliation(s)
- Srinivas Ramachandran
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7260, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Vrolix K, Fraussen J, Molenaar PC, Losen M, Somers V, Stinissen P, De Baets MH, Martínez-Martínez P. The auto-antigen repertoire in myasthenia gravis. Autoimmunity 2010; 43:380-400. [PMID: 20380581 DOI: 10.3109/08916930903518073] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Myasthenia Gravis (MG) is an antibody-mediated autoimmune disorder affecting the postsynaptic membrane of the neuromuscular junction (NMJ). MG is characterized by an impaired signal transmission between the motor neuron and the skeletal muscle cell, caused by auto-antibodies directed against NMJ proteins. The auto-antibodies target the nicotinic acetylcholine receptor (nAChR) in about 90% of MG patients. In approximately 5% of MG patients, the muscle specific kinase (MuSK) is the auto-antigen. In the remaining 5% of MG patients, however, antibodies against the nAChR or MuSK are not detectable (idiopathic MG, iMG). Although only the anti-nAChR and anti-MuSK auto-antibodies have been demonstrated to be pathogenic, several other antibodies recognizing self-antigens can also be found in MG patients. Various auto-antibodies associated with thymic abnormalities have been reported, as well as many non-MG-specific auto-antibodies. However, their contribution to the cause, pathology and severity of the disease is still poorly understood. Here, we comprehensively review the reported auto-antibodies in MG patients and discuss their role in the pathology of this autoimmune disease.
Collapse
Affiliation(s)
- Kathleen Vrolix
- Division of Neuroscience, School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
5
|
|
6
|
Abstract
The ryanodine receptors (RyRs) are a family of Ca2+ release channels found on intracellular Ca2+ storage/release organelles. The RyR channels are ubiquitously expressed in many types of cells and participate in a variety of important Ca2+ signaling phenomena (neurotransmission, secretion, etc.). In striated muscle, the RyR channels represent the primary pathway for Ca2+ release during the excitation-contraction coupling process. In general, the signals that activate the RyR channels are known (e.g., sarcolemmal Ca2+ influx or depolarization), but the specific mechanisms involved are still being debated. The signals that modulate and/or turn off the RyR channels remain ambiguous and the mechanisms involved unclear. Over the last decade, studies of RyR-mediated Ca2+ release have taken many forms and have steadily advanced our knowledge. This robust field, however, is not without controversial ideas and contradictory results. Controversies surrounding the complex Ca2+ regulation of single RyR channels receive particular attention here. In addition, a large body of information is synthesized into a focused perspective of single RyR channel function. The present status of the single RyR channel field and its likely future directions are also discussed.
Collapse
Affiliation(s)
- Michael Fill
- Department of Physiology, Loyola University Chicago, Maywood, Illinois 60153, USA
| | | |
Collapse
|
7
|
Kennedy HJ, Meech RW. Fast Ca2+ signals at mouse inner hair cell synapse: a role for Ca2+-induced Ca2+ release. J Physiol 2002; 539:15-23. [PMID: 11850498 PMCID: PMC2290124 DOI: 10.1113/jphysiol.2001.013171] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Inner hair cells of the mammalian cochlea translate acoustic stimuli into 'phase-locked' nerve impulses with frequencies of up to at least 1 kHz. Little is known about the intracellular Ca2+ signal that links transduction to the release of neurotransmitter at the afferent synapse. Here, we use confocal microscopy to provide evidence that Ca2+-induced Ca2+ release (CICR) may contribute to the mechanism. Line scan images (2 ms repetition rate) of neonatal mouse inner hair cells filled with the fluorescent indicator FLUO-3, revealed a transient increase in intracellular Ca2+ concentration ([Ca2+]i) during brief (5-50 ms) depolarizing commands under voltage clamp. The amplitude of the [Ca2+]i transient depended upon the Ca2+ concentration in the bathing medium in the range 0-1.3 mM. [Ca2+]i transients were confined to a region near the plasma membrane at the base of the cell in the vicinity of the afferent synapses. The change in [Ca2+]i appeared uniform throughout the entire basal sub-membrane space and we were unable to observe hotspots of activity. Both the amplitude and the rate of rise of the [Ca2+]i transient was reduced by external ryanodine (20 microM), an agent that blocks Ca2+ release from the endoplasmic reticulum. Intracellular Cs+, commonly used to record at presynaptic sites, produced a similar effect. We conclude that both ryanodine and intracellular Cs+ block CICR in inner hair cells. We discuss the contribution of CICR to the measured [Ca2+]i transient, the implications for synaptic transmission at the afferent synapse and the significance of its sensitivity to intracellular Cs+.
Collapse
Affiliation(s)
- Helen J Kennedy
- Department of Physiology, University of Bristol, Bristol, BS8 1TD, UK.
| | | |
Collapse
|
8
|
Gathercole DV, Colling DJ, Skepper JN, Takagishi Y, Levi AJ, Severs NJ. Immunogold-labeled L-type calcium channels are clustered in the surface plasma membrane overlying junctional sarcoplasmic reticulum in guinea-pig myocytes-implications for excitation-contraction coupling in cardiac muscle. J Mol Cell Cardiol 2000; 32:1981-94. [PMID: 11040103 DOI: 10.1006/jmcc.2000.1230] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ca(2+) release through ryanodine receptors, located in the membrane of the junctional sarcoplasmic reticulum (SR), initiates contraction of cardiac muscle. Ca(2+)influx through plasma membrane L-type Ca(2+)channels is thought to be an important trigger for opening ryanodine receptors ("Ca(2+)-induced Ca(2+)-release"). Optimal transmission of the transmembrane Ca(2+)influx signal to SR release is predicted to involve spatial juxtaposition of L-type Ca(2+)channels to the ryanodine receptors of the junctional SR. Although such spatial coupling has often been implicitly assumed, and data from immunofluorescence microscopy are consistent with its existence, the definitive demonstration of such a structural organization in mammalian tissue is lacking at the electron-microscopic level. To determine the spatial distribution of plasma membrane L-type Ca(2+)channels and their location in relation to underlying junctional SR, we applied two high-resolution immunogold-labeling techniques, label-fracture and cryothin-sectioning, combined with quantitative analysis, to guinea-pig ventricular myocytes. Label-fracture enabled visualization of colloidal gold-labeled L-type Ca(2+)channels in planar freeze-fracture electron-microscopic views of the plasma membrane. Mathematical analysis of the gold label distribution (by nearest-neighbor distance distribution and the radial distribution function) demonstrated genuine clustering of the labeled channels. Gold-labeled cryosections showed that labeled L-type Ca(2+)channels quantitatively predominated in domains of the plasma membrane overlying junctional SR. These findings provide an ultrastructural basis for functional coupling between L-type Ca(2+)channels and junctional SR and for excitation-contraction coupling in guinea-pig cardiac muscle.
Collapse
Affiliation(s)
- D V Gathercole
- National Heart and Lung Institute, Imperial College of Science, Technology and Medicine, London, UK
| | | | | | | | | | | |
Collapse
|
9
|
Sharma C, Smith T, Li S, Schroepfer GJ, Needleman DH. Inhibition of Ca2+ release channel (ryanodine receptor) activity by sphingolipid bases: mechanism of action. Chem Phys Lipids 2000; 104:1-11. [PMID: 10660207 DOI: 10.1016/s0009-3084(99)00106-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sphingosine inhibits the activity of the skeletal muscle Ca2+ release channel (ryanodine receptor) and is a noncompetitive inhibitor of [3H]ryanodine binding (Needleman et al., Am. J. Physiol. 272, C1465-1474, 1997). To determine the contribution of other sphingolipids to the regulation of ryanodine receptor activity, several sphingolipid bases were assessed for their ability to alter [3H]ryanodine binding to sarcoplasmic reticulum (SR) membranes and to modulate the activity of the Ca2+ release channel. Three lipids, N,N-dimethylsphingosine, dihydrosphingosine, and phytosphingosine, inhibited [3H]ryanodine binding to both skeletal and cardiac SR membranes. However, the potency of these three lipids and sphingosine was lower in rabbit cardiac membranes when compared to rabbit skeletal muscle membranes and when compared to sphingosine. Like sphingosine, the lipids inhibited [3H]ryanodine binding by greatly increasing the rate of dissociation of bound [3H]ryanodine from SR membranes, indicating that these three sphingolipid bases were noncompetitive inhibitors of [3H]ryanodine binding. These bases also decreased the activity of the Ca2+ release channel incorporated into planar lipid bilayers by stabilizing a long closed state. Sphingosine-1-PO4 and C6 to C18 ceramides of sphingosine had no significant effect on [3H]ryanodine binding to cardiac or skeletal muscle SR membranes. Saturation of the double bond at positions 4-5 decreased the ability of the sphingolipid bases to inhibit [3H]ryanodine binding 2-3 fold compared to sphingosine. In summary, our data indicate that other endogenous sphingolipid bases are capable of modulating the activity of the Ca2+ release channel and as a class possess a common mechanism of inhibition.
Collapse
Affiliation(s)
- C Sharma
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
10
|
Suko J, Drobny H, Hellmann G. Activation and inhibition of purified skeletal muscle calcium release channel by NO donors in single channel current recordings. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1451:271-87. [PMID: 10556582 DOI: 10.1016/s0167-4889(99)00098-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The actions of the nitric oxide (NO) donors 1-hydroxy-2-oxo-3-(N-methyl-3-aminopropyl)-3 methyl-1-triazine (NOC-7), S-nitrosoacetylcysteine (CySNO) and S-nitrosoglutathione (GSNO) on the purified calcium release channel (ryanodine receptor) of rabbit skeletal muscle were determined by single channel current recordings. In addition, the activation of the NO donor modulated calcium release channel by the sulfhydryl oxidizing organic mercurial compound 4-(chloromercuri)phenylsulfonic acid (4-CMPS) was investigated. NOC-7 (0.1 and 0.3 mM) and CySNO (0.4 and 0.8 mM) increased the open probability (P(o)) of the calcium release channel at activating calcium concentrations (20-100 microM Ca(2+)) by 60-100%, with no effect on the current amplitude; this activation was abolished by the specific sulfhydryl reducing agent DTT. High concentrations of CySNO (1.6-2 mM) decreased P(o). Activation by GSNO (1 mM) was observed in two thirds of the experiments, but 2 mM and 4 mM GSNO markedly reduced P(o) at activating Ca(2+) (20-100 microM). In contrast to 4-CMPS, NOC-7 or GSNO had no effect at subactivating free Ca(2+) (0.6 microM). 4-CMPS further increased the open probability of NOC-7- or CySNO-stimulated channels and reversed transiently the reduced open probability of CySNO or GSNO inhibited channels at activating free Ca(2+). High concentrations of GSNO did not prevent channel activation of 4-CMPS at subactivating free Ca(2+). The NOC-7-, CySNO- or GSNO-modified channels were completely blocked by ruthenium red. It is suggested that nitrosylation/oxidation of sulfhydryls by NO donors and oxidation of sulfhydryls by 4-CMPS affect different cysteine residues essential in the gating of the calcium release channel.
Collapse
Affiliation(s)
- J Suko
- Institute of Pharmacology, University of Vienna, Waehringerstrasse 13a, A-1090, Vienna, Austria
| | | | | |
Collapse
|
11
|
Bolton TB, Prestwich SA, Zholos AV, Gordienko DV. Excitation-contraction coupling in gastrointestinal and other smooth muscles. Annu Rev Physiol 1999; 61:85-115. [PMID: 10099683 DOI: 10.1146/annurev.physiol.61.1.85] [Citation(s) in RCA: 179] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The main contributors to increases in [Ca2+]i and tension are the entry of Ca2+ through voltage-dependent channels opened by depolarization or during action potential (AP) or slow-wave discharge, and Ca2+ release from store sites in the cell by the action of IP3 or by Ca(2+)-induced Ca(2+)-release (CICR). The entry of Ca2+ during an AP triggers CICR from up to 20 or more subplasmalemmal store sites (seen as hot spots, using fluorescent indicators); Ca2+ waves then spread from these hot spots, which results in a rise in [Ca2+]i throughout the cell. Spontaneous transient releases of store Ca2+, previously detected as spontaneous transient outward currents (STOCs), are seen as sparks when fluorescent indicators are used. Sparks occur at certain preferred locations--frequent discharge sites (FDSs)--and these and hot spots may represent aggregations of sarcoplasmic reticulum scattered throughout the cytoplasm. Activation of receptors for excitatory signal molecules generally depolarizes the cell while it increases the production of IP3 (causing calcium store release) and diacylglycerols (which activate protein kinases). Activation of receptors for inhibitory signal molecules increases the activity of protein kinases through increases in cAMP or cGMP and often hyperpolarizes the cell. Other receptors link to tyrosine kinases, which trigger signal cascades interacting with trimeric G-protein systems.
Collapse
Affiliation(s)
- T B Bolton
- Department of Pharmacology and Clinical Pharmacology, St George's Hospital Medical School, London, United Kingdom.
| | | | | | | |
Collapse
|
12
|
Chen DP, Xu L, Tripathy A, Meissner G, Eisenberg B. Selectivity and permeation in calcium release channel of cardiac muscle: alkali metal ions. Biophys J 1999; 76:1346-66. [PMID: 10049318 PMCID: PMC1300114 DOI: 10.1016/s0006-3495(99)77297-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Current was measured from single open channels of the calcium release channel (CRC) of cardiac sarcoplasmic reticulum (over the range +/-180 mV) in pure and mixed solutions (e.g., biionic conditions) of the alkali metal ions Li+, K+, Na+, Rb+, Cs+, ranging in concentration from 25 mM to 2 M. The current-voltage (I-V) relations were analyzed by an extension of the Poisson-Nernst-Planck (PNP) formulation of electrodiffusion, which includes local chemical interaction described by an offset in chemical potential, which likely reflects the difference in dehydration/solvation/rehydration energies in the entry/exit steps of permeation. The theory fits all of the data with few adjustable parameters: the diffusion coefficient of each ion species, the average effective charge distribution on the wall of the pore, and an offset in chemical potential for lithium and sodium ions. In particular, the theory explains the discrepancy between "selectivities" defined by conductance sequence and "selectivities" determined by the permeability ratios (i.e., reversal potentials) in biionic conditions. The extended PNP formulation seems to offer a successful combined treatment of selectivity and permeation. Conductance selectivity in this channel arises mostly from friction: different species of ions have different diffusion coefficients in the channel. Permeability selectivity of an ion is determined by its electrochemical potential gradient and local chemical interaction with the channel. Neither selectivity (in CRC) seems to involve different electrostatic interaction of different ions with the channel protein, even though the ions have widely varying diameters.
Collapse
Affiliation(s)
- D P Chen
- Department of Molecular Biophysics and Physiology, Rush Medical College, Chicago, Illinois 60612, USA.
| | | | | | | | | |
Collapse
|
13
|
Zhang JJ, Williams AJ, Sitsapesan R. Evidence for novel caffeine and Ca2+ binding sites on the lobster skeletal ryanodine receptor. Br J Pharmacol 1999; 126:1066-74. [PMID: 10193789 PMCID: PMC1571222 DOI: 10.1038/sj.bjp.0702400] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/1998] [Revised: 11/19/1998] [Accepted: 12/02/1998] [Indexed: 11/09/2022] Open
Abstract
1. The effects of Ca2+, ATP and caffeine on the gating of lobster skeletal muscle ryanodine receptors (RyR) was investigated after reconstitution of the channels into planar phospholipid bilayers and by using [3H]-ryanodine binding studies. 2. The single channel studies reveal that the EC50 (60 microM) for activation of the lobster skeletal RyR by Ca2+ as the sole ligand is higher than for any other isoform of RyR studied. 3. Inactivation of the channel by Ca2+ (EC50 = 1 mM) occurs at concentrations slightly higher than those required to inactivate mammalian skeletal RyR (RyR1) but lower than those required to inactivate mammalian cardiac RyR (RyR2). 4. Lifetime analysis demonstrates that cytosolic Ca2+, as the sole activating ligand, cannot fully open the lobster skeletal RyR (maximum Po approximately 0.2). The mechanism for the increase in open probability (Po) is an increase in both the frequency and the duration of the open events. 5. ATP is a very effective activator of the lobster RyR and can almost fully open the channel in the presence of activating cytosolic [Ca2+]. In the presence of 700 microM Ca2+, 1 mM ATP increased Po to approximately 0.8. 6. Caffeine, often used as a tool to identify the presence of RyR channels, is relatively ineffective and cannot increase Po above the level that can be attained with Ca2+ alone. 7. The results reveal that caffeine increases Po by a different mechanism to that of cytosolic Ca2+ demonstrating that the mechanism for channel activation by caffeine is not 'sensitization' to cytosolic Ca2+. 8. By studying the mechanisms involved in the activation of the lobster RyR we have demonstrated that the channel responds in a unique manner to Ca2+ and to caffeine. The results strongly indicate that these ligand binding sites on the channel are different to those on mammalian isoforms of RyR.
Collapse
Affiliation(s)
- J J Zhang
- Cardiac Medicine, Imperial College School of Medicine, London, England, UK
| | | | | |
Collapse
|
14
|
Ondrias K, Marx SO, Gaburjakova M, Marks AR. FKBP12 modulates gating of the ryanodine receptor/calcium release channel. Ann N Y Acad Sci 1998; 853:149-56. [PMID: 10603943 DOI: 10.1111/j.1749-6632.1998.tb08263.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Excitation-contraction (EC) coupling in muscle requires the activation of intracellular calcium release channels (CRC). Four type 1 ryanodine receptor (RyR1) molecules form each tetrameric CRC. Each RyR1 contains a binding site for the FK506 binding protein (FKBP12), a cis-trans peptidyl-prolyl isomerase that is required for coordinated gating of the four RyR1 subunits comprising the channel. When FKBP12 is bound to RyR1, it stabilizes the four subunits that form each CRC. We propose that binding of one FKBP12 to each RyR1 lowers the energy of twisted-amide peptidyl-prolyl bonds and stabilizes RyR1 in a conformation that permits coordinated gating of the four RyR1 subunits.
Collapse
Affiliation(s)
- K Ondrias
- Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | | | | | | |
Collapse
|
15
|
Suko J, Hellmann G. Modification of sulfhydryls of the skeletal muscle calcium release channel by organic mercurial compounds alters Ca2+ affinity of regulatory Ca2+ sites in single channel recordings and [3H]ryanodine binding. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1404:435-50. [PMID: 9739172 DOI: 10.1016/s0167-4889(98)00075-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The actions of two organic mercurial compounds, 4-(chloromercuri)phenyl-sulfonic acid (4-CMPS) and p-chloromercuribenzoic acid (p-CMB) on the calcium release channel (ryanodine receptor) from rabbit skeletal muscle were determined by single channel recordings with the purified calcium release channel, radioligand binding to sarcoplasmic reticulum vesicles (HSR) and calcium release from HSR. p-CMB or 4-CMPS (20-100 microM) increased the mean open probability (Po) of the calcium channel at subactivating (20 nM), maximally activating (20-100 microM and inhibitory (1-4 mM) Ca2+ concentrations, with no effect on unitary conductance. This activation was partly reversed by 2 mM DTT. Both compounds affected the channels only from the cytosolic side, but not from the trans side. 100 microM 4-CMPS caused a transient increase in Po, followed by a low activity state within 1 min. At inhibitory Ca2+ concentrations Po was increased to values observed with maximally activating Ca2+ or lower, inhibitory Ca2+ concentrations. The p-CMB/4-CMPS modified channels were ryanodine sensitive and blocked by ruthenium red. [3H]Ryanodine binding was increased up to four-fold with 3-15 microM 4-CMPS/p-CMB (Hill coefficient 1.7-2.0) at 4 microM Ca2+ and reduced at high concentrations (50-200 microM). The increase in [3H]ryanodine binding by 10 microM 4-CMPS was completely inhibited by 2 mM DTT. 4-CMPS significantly increased the affinity for the high affinity calcium activation sites and decreased the affinity of low affinity calcium inhibitory sites of specific [3H]ryanodine binding. 4-CMPS increased the affinity of the ryanodine receptor for high affinity ryanodine binding without a change in receptor density. 4-CMPS induced a rapid, concentration-dependent, biphasic calcium release from passively calcium-loaded HSR vesicles at subactivating Ca2+ concentrations (20 nM), which was partly inhibited by 4 mM DTT and completely blocked by 20 microM ruthenium red. It is suggested that the 4-CMPS-induced modulation of essential sulfhydryls involved in the gating of the calcium release channel results in a modulation of the apparent calcium affinity of the activating high affinity and inhibitory low affinity calcium binding sites of the calcium release channel.
Collapse
Affiliation(s)
- J Suko
- Institute of Pharmacology, University of Vienna, Waehringerstrasse 13a, A-1090 Vienna, Austria
| | | |
Collapse
|
16
|
Shoshan-Barmatz V, Ashley RH. The structure, function, and cellular regulation of ryanodine-sensitive Ca2+ release channels. INTERNATIONAL REVIEW OF CYTOLOGY 1998; 183:185-270. [PMID: 9666568 DOI: 10.1016/s0074-7696(08)60145-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The fundamental biological process of Ca2+ signaling is known to be important in most eukaryotic cells, and inositol 1,2,5-trisphosphate and ryanodine receptors, intracellular Ca2+ release channels encoded by two distantly related gene families, are central to this phenomenon. Ryanodine receptors in the sarcoplasmic reticulum of skeletal and cardiac muscle have a predominant role in excitation-contraction coupling, but the channels are also present in the endoplasmic reticulum of noncontractile tissues including the central nervous system and the immune system. In all, three highly homologous ryanodine receptor isoforms have been identified, all very large proteins which assemble as (homo)tetramers of approximately 2 MDa. They contain large cytoplasmically disposed regulatory domains and are always associated with other structural or regulatory proteins, including calmodulin and immunophilins, which can have marked effects on channel function. The type 1 isoform in skeletal muscle is electromechanically coupled to surface membrane voltage sensors, whereas the remaining isoforms appear to be activated solely by endogenous cytoplasmic second messengers or other ligands, including Ca2+ itself ("Ca(2+)-induced Ca2+ release"). This review concentrates on ryanodine receptor structure-function relationships as probed by a variety of methods and on the molecular mechanisms of channel modulation at the cellular level (including evidence for the regulation of gene expression and transcription). It also touches on the relevance of ryanodine receptors to complex cellular functions and disease.
Collapse
Affiliation(s)
- V Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | | |
Collapse
|
17
|
Mak DO, Foskett JK. Effects of divalent cations on single-channel conduction properties of Xenopus IP3 receptor. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:C179-88. [PMID: 9688849 DOI: 10.1152/ajpcell.1998.275.1.c179] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The effects of Mg2+ and Ba2+ on single-channel properties of the inositol 1,4,5-trisphosphate receptor (IP3R) were studied by patch clamp of isolated nuclei from Xenopus oocytes. In 140 mM K+ the IP3R channel kinetics and presence of conductance substates were similar over a range (0-9.5 mM) of free Mg2+. In 0 mM Mg2+ the channel current-voltage (I-V) relation was linear with conductance of approximately 320 pS. Conductance varied slowly and continuously over a wide range (SD approximately 60 pS) and sometimes fluctuated during single openings. The presence of Mg2+ on either or both sides of the channel reduced the current (blocking constant approximately 0.6 mM in symmetrical Mg2+), as well as the range of conductances observed, and made the I-V relation nonlinear (slope conductance approximately 120 pS near 0 mV and approximately 360 pS at +/-70 mV in symmetrical 2.5 mM Mg2+). Ba2+ exhibited similar effects on channel conductance. Mg2+ and Ba2+ permeated the channel with a ratio of permeability of Ba2+ to Mg2+ to K+ of 3.5:2.6:1. These results indicate that divalent cations induce nonlinearity in the I-V relation and reduce current by a mechanism involving permeation block of the IP3R due to strong binding to site(s) in the conduction pathway. Furthermore, stabilization of conductance by divalent cations reveals a novel interaction between the cations and the IP3R.
Collapse
Affiliation(s)
- D O Mak
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6100, USA
| | | |
Collapse
|
18
|
Stephenson DG, Lamb GD, Stephenson GM. Events of the excitation-contraction-relaxation (E-C-R) cycle in fast- and slow-twitch mammalian muscle fibres relevant to muscle fatigue. ACTA PHYSIOLOGICA SCANDINAVICA 1998; 162:229-45. [PMID: 9578368 DOI: 10.1046/j.1365-201x.1998.0304f.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The excitation-contraction-relaxation cycle (E-C-R) in the mammalian twitch muscle comprises the following major events: (1) initiation and propagation of an action potential along the sarcolemma and transverse (T)-tubular system; (2) detection of the T-system depolarization signal and signal transmission from the T-tubule to the sarcoplasmic reticulum (SR) membrane; (3) Ca2+ release from the SR; (4) transient rise of myoplasmic [Ca2+]; (5) transient activation of the Ca2+-regulatory system and of the contractile apparatus; (6) Ca2+ reuptake by the SR Ca2+ pump and Ca2+ binding to myoplasmic sites. There are many steps in the E-C-R cycle which can be seen as potential sites for muscle fatigue and this review explores how structural and functional differences between the fast- and slow-twitch fibres with respect to the E-C-R cycle events can explain to a great extent differences in their fatiguability profiles.
Collapse
Affiliation(s)
- D G Stephenson
- School of Zoology, La Trobe University, Bundoora, Victoria, Australia
| | | | | |
Collapse
|
19
|
Laver DR, Peter WG. Interpretation of substates in ion channels: unipores or multipores? PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 1998; 67:99-140. [PMID: 9446932 DOI: 10.1016/s0079-6107(97)00008-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- D R Laver
- John Curtin School of Medical Research, Australian National University, Camberra, ACT, Australia
| | | |
Collapse
|
20
|
Tameyasu T. Regulation of Contraction in Heart Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1998. [DOI: 10.1007/978-1-4684-6039-1_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Chen D, Xu L, Tripathy A, Meissner G, Eisenberg B. Permeation through the calcium release channel of cardiac muscle. Biophys J 1997; 73:1337-54. [PMID: 9284302 PMCID: PMC1181034 DOI: 10.1016/s0006-3495(97)78167-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Current voltage (I-V) relations were measured from the calcium release channel (CRC) of the sarcoplasmic reticulum of cardiac muscle in 12 KCl solutions, symmetrical and asymmetrical, from 25 mM to 2 M. I-V curves are nearly linear, in the voltage range +/- 150 mV approximately 12kT/e, even in asymmetrical solutions, e.g., 2 M // 100 mM. It is awkward to describe straight lines as sums of exponentials in a wide range of solutions and potentials, and so traditional barrier models have difficulty fitting this data. Diffusion theories with constant fields predict curvilinear I-V relations, and so they are also unsatisfactory. The Poisson and Nernst-Planck equations (PNP) form a diffusion theory with variable fields. They fit the data by using adjustable parameters for the diffusion constant of each ion and for the effective density of fixed (i.e., permanent) charge P(x) along the channel's "filter" (7-A diameter, 10 A long). If P(x) is described by just one parameter, independent of x (i.e., P(x) = P0 = -4.2 M), the fits are satisfactory (RMS error/RMS current = 6.4/67), and the estimates of diffusion coefficients are reasonable D(K) = 1.3 x 10(-6) cm2/s, D(Cl) = 3.9 x 10(-6) cm2/s. The CRC seems to have a small selectivity filter with a very high density of permanent charge. This may be a design principle of channels specialized for large flux. The Appendix derives barrier models, and their prefactor, from diffusion theories (with variable fields) and argues that barrier models are poor descriptions of CRCs in particular and open channels in general.
Collapse
Affiliation(s)
- D Chen
- Department of Molecular Biophysics and Physiology, Rush Medical College, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
22
|
Levi AJ, Dalton GR, Hancox JC, Mitcheson JS, Issberner J, Bates JA, Evans SJ, Howarth FC, Hobai IA, Jones JV. Role of intracellular sodium overload in the genesis of cardiac arrhythmias. J Cardiovasc Electrophysiol 1997; 8:700-21. [PMID: 9209972 DOI: 10.1111/j.1540-8167.1997.tb01834.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A number of clinical cardiac disorders may be associated with a rise of the intracellular Na concentration (Na(i)) in heart muscle. A clear example is digitalis toxicity, in which excessive inhibition of the Na/K pump causes the Na(i) concentration to become raised above the normal level. Especially in digitalis toxicity, but also in many other situations, the rise of Na(i) may be an important (or contributory) cause of increased cardiac arrhythmias. In this review, we consider the mechanisms by which a raised Na(i) may cause cardiac arrhythmias. First, we describe the factors that regulate Na(i), and we demonstrate that the equilibrium level of Na(i) is determined by a balance between Na entry into the cell, and Na extrusion from the cell. A number of mechanisms are responsible for Na entry into the cell, whereas the Na/K pump appears to be the main mechanism for Na extrusion. We then consider the processes by which an increased level of Nai might contribute to cardiac arrhythmias. A rise of Na(i) is well known to result in an increase of intracellular Ca, via the important and influential Na/Ca exchange mechanism in the cell membrane of cardiac muscle cells. A rise of intracellular Ca modulates the activity of a number of sarcolemmal ion channels and affects release of intracellular Ca from the sarcoplasmic reticulum, all of which might be involved in causing arrhythmia. It is possible that the increase in contractile force that results from the rise of intracellular Ca may initiate or exacerbate arrhythmia, since this will increase wall stress and energy demands in the ventricle, and an increase in wall stress may be arrhythmogenic. In addition, the rise of Na(i) is anticipated to modulate directly a number of ion channels and to affect the regulation of intracellular pH, which also may be involved in causing arrhythmia. We also present experiments in this review, carried out on the working rat heart preparation, which suggest that a rise of Na(i) causes an increase of wall stress-induced arrhythmia in this model. In addition, we have investigated the effect on wall stress-induced arrhythmia of maneuvers that might be anticipated to change intracellular Ca, and this has allowed identification of some of the factors involved in causing arrhythmia in the working rat heart.
Collapse
Affiliation(s)
- A J Levi
- Department of Physiology, School of Medical Sciences, University of Bristol, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Mak DO, Foskett JK. Single-channel kinetics, inactivation, and spatial distribution of inositol trisphosphate (IP3) receptors in Xenopus oocyte nucleus. J Gen Physiol 1997; 109:571-87. [PMID: 9154905 PMCID: PMC2217068 DOI: 10.1085/jgp.109.5.571] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/1996] [Accepted: 02/24/1997] [Indexed: 02/04/2023] Open
Abstract
Single-channel properties of the Xenopus inositol trisphosphate receptor (IP3R) ion channel were examined by patch clamp electrophysiology of the outer nuclear membrane of isolated oocyte nuclei. With 140 mM K+ as the charge carrier (cytoplasmic [IP3] = 10 microM, free [Ca2+] = 200 nM), the IP3R exhibited four and possibly five conductance states. The conductance of the most-frequently observed state M was 113 pS around 0 mV and approximately 300 pS at 60 mV. The channel was frequently observed with high open probability (mean P(o) = 0.4 at 20 mV). Dwell time distribution analysis revealed at least two kinetic states of M with time constants tau < 5 ms and approximately 20 ms; and at least three closed states with tau approximately 1 ms, approximately 10 ms, and >1 s. Higher cytoplasmic potential increased the relative frequency and tau of the longest closed state. A novel "flicker" kinetic mode was observed, in which the channel alternated rapidly between two new conductance states: F1 and F2. The relative occupation probability of the flicker states exhibited voltage dependence described by a Boltzmann distribution corresponding to 1.33 electron charges moving across the entire electric field during F1 to F2 transitions. Channel run-down or inactivation (tau approximately 30 s) was consistently observed in the continuous presence of IP3 and the absence of change in [Ca2+]. Some (approximately 10%) channel disappearances could be reversed by an increase in voltage before irreversible inactivation. A model for voltage-dependent channel gating is proposed in which one mechanism controls channel opening in both the normal and flicker modes, whereas a separate independent mechanism generates flicker activity and voltage-reversible inactivation. Mapping of functional channels indicates that the IP3R tends to aggregate into microscopic (<1 microm) as well as macroscopic (approximately 10 microm) clusters. Ca2+-independent inactivation of IP3R and channel clustering may contribute to complex [Ca2+] signals in cells.
Collapse
Affiliation(s)
- D O Mak
- Department of Physiology, University of Pennsylvania, Philadelphia 19104-6100, USA.
| | | |
Collapse
|
24
|
Hernández-Cruz A, Escobar AL, Jiménez N. Ca(2+)-induced Ca2+ release phenomena in mammalian sympathetic neurons are critically dependent on the rate of rise of trigger Ca2+. J Gen Physiol 1997; 109:147-67. [PMID: 9041445 PMCID: PMC2220057 DOI: 10.1085/jgp.109.2.147] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/1996] [Accepted: 11/14/1996] [Indexed: 02/03/2023] Open
Abstract
The role of ryanodine-sensitive intracellular Ca2+ stores present in nonmuscular cells is not yet completely understood. Here we examine the physiological parameters determining the dynamics of caffeine-induced Ca2+ release in individual fura 2-loaded sympathetic neurons. Two ryanodine-sensitive release components were distinguished: an early, transient release (TR) and a delayed, persistent release (PR). The TR components shows refractoriness, depends on the filling status of the store, and requires caffeine concentrations > or = 10 mM. Furthermore, it is selectively suppressed by tetracaine and intracellular BAPTA, which interfere with Ca(2+)-mediated feedback loops, suggesting that it constitutes a Ca(2+)-induced Ca(2+)-release phenomenon. The dynamics of release is markedly affected when Sr2+ substitutes for Ca2+, indicating that Sr2+ release may operate with lower feedback gain than Ca2+ release. Our data indicate that when the initial release occurs at an adequately fast rate, Ca2+ triggers further release, producing a regenerative response, which is interrupted by depletion of releasable Ca2+ and Ca(2+)-dependent inactivation. A compartmentalized linear diffusion model can reproduce caffeine responses: When the Ca2+ reservoir is full, the rapid initial Ca2+ rise determines a faster occupation of the ryanodine receptor Ca2+ activation site giving rise to a regenerative release. With the store only partially loaded, the slower initial Ca2+ rise allows the inactivating site of the release channel to become occupied nearly as quickly as the activating site, thereby suppressing the initial fast release. The PR component is less dependent on the store's Ca2+ content. This study suggests that transmembrane Ca2+ influx in rat sympathetic neurons does not evoke widespread amplification by CICR because of its inability to raise [Ca2+] near the Ca2+ release channels sufficiently fast to overcome their Ca(2+)-dependent inactivation. Conversely, caffeine-induced Ca2+ release can undergo considerable amplification especially when Ca2+ stores are full. We propose that the primary function of ryanodine-sensitive stores in neurons and perhaps in other nonmuscular cells, is to emphasize subcellular Ca2+ gradients resulting from agonist-induced intracellular release. The amplification gain is dependent both on the agonist concentration and on the filling status of intracellular Ca2+ stores.
Collapse
Affiliation(s)
- A Hernández-Cruz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria México City, D.F. México.
| | | | | |
Collapse
|
25
|
Tunwell RE, Wickenden C, Bertrand BM, Shevchenko VI, Walsh MB, Allen PD, Lai FA. The human cardiac muscle ryanodine receptor-calcium release channel: identification, primary structure and topological analysis. Biochem J 1996; 318 ( Pt 2):477-87. [PMID: 8809036 PMCID: PMC1217646 DOI: 10.1042/bj3180477] [Citation(s) in RCA: 121] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Rapid Ca2+ efflux from intracellular stores during cardiac muscle excitation-contraction coupling is mediated by the ryanodine-sensitive calcium-release channel, a large homotetrameric complex present in the sarcoplasmic reticulum. We report here the identification, primary structure and topological analysis of the ryanodine receptor-calcium release channel from human cardiac muscle (hRyR-2). Consistent with sedimentation and immunoblotting studies on the hRyR-2 protein, sequence analysis of ten overlapping cDNA clones reveals an open reading frame of 14901 nucleotides encoding a protein of 4967 amino acid residues with a predicted molecular mass of 564 569 Da for hRyR-2. In-frame insertions corresponding to eight and ten amino acid residues were found in two of the ten cDNAs isolated, suggesting that novel, alternatively spliced transcripts of the hRyR-2 gene might exist. Six hydrophobic stretches, which are present within the hRyR-2 C-terminal 500 amino acids and are conserved in all RyR sequences, may be involved in forming the transmembrane domain that constitutes the Ca(2+)-conducting pathway, in agreement with competitive ELISA studies with a RyR-2-specific antibody. Sequence alignment of hRyR-2 with other RyR isoforms indicates a high level of overall identity within the RyR family, with the exception of two important regions that exhibit substantial variability. Phylogenetic analysis suggests that the RyR-2 isoform diverged from a single ancestral gene before the RyR-1 and RyR-3 isoforms to form a distinct branch of the RyR family tree.
Collapse
Affiliation(s)
- R E Tunwell
- MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, U.K
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Spontaneous transient outward currents (STOCs) lasting about 100 ms occur in single smooth muscle cells and represent the simultaneous opening of up to a hundred calcium-activated potassium (BK) channels. The recent observation of brief focal releases of sarcoplasmic reticulum (SR) calcium ('sparks') in smooth muscle cells has provided support for the original suggestion that STOCs arise due to the spontaneous releases of calcium from the SR close to the sarcolemma. However, it is possible that such releases occur in a region of close apposition of SR membrane and sarcolemma about 0.1 microns wide ('junctional space') in which case they would be detectable by endogenous calcium-sensitive molecules such as BK channels but, using present confocal microscopy technique, not by calcium-indicator dyes introduced into the cell; should calcium escape from the junctional space then it may be visualised as 'sparks' by the fluorescent emission from calcium-indicator dyes using confocal microscopy. Some STOCs seem too large to represent the effect of a single 'spark' and some form of calcium-induced calcium release or 'macrospark' may be involved in their generation. Depletion of calcium stores by caffeine, ryanodine, or by activation of receptors linked to the phospholipase C/inositol trisphosphate system abolishes STOCs. However, low concentrations of caffeine or inositol trisphosphate accelerate STOC discharge by an unknown mechanism and often decrease STOC size presumably by depleting store calcium; similar effects are produced by agents such as cyclopiazonic acid and thapsigargin which inhibit calcium storage mechanisms (largely the SR calcium pump).
Collapse
Affiliation(s)
- T B Bolton
- Department of Pharmacology and Clinical Pharmacology, St George's Hospital Medical School, London, UK.
| | | |
Collapse
|
27
|
Levi AJ, Li J, Spitzer KW, Bridge JH. Effect on the indo-1 transient of applying Ca2+ channel blocker for a single beat in voltage-clamped guinea-pig cardiac myocytes. J Physiol 1996; 494 ( Pt 3):653-73. [PMID: 8865064 PMCID: PMC1160667 DOI: 10.1113/jphysiol.1996.sp021522] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
1. We used rapid solution changes to investigate the mechanisms which trigger Ca2+ release from the sarcoplasmic reticulum (SR) in guinea-pig ventricular myocytes. We patch-clamped myocytes at 36 degrees C and used indo-1 to monitor intracellular Ca2+. Before each test pulse, we established a standard level of SR Ca2+ load by applying a train of conditioning pulses. 2. We switched rapidly to 32 microM nifedipine (an L-type Ca2+ current (ICa,L) blocker) 8 s before a test pulse, and just after applying nifedipine we applied a ramp depolarization to pre-block Ca2+ channels. We found that ICa,L elicited by the following test pulse was inhibited almost completely (98-99% inhibition). 3. The indo-1 transient elicited by an 800 ms depolarizing pulse showed a rapid initial rise which was inhibited by ryanodine-thapsigargin. This indicated that the rapid rise was due to Ca2+ release from the SR, and therefore provides an index of SR Ca2+ release. 4. In cells dialysed internally with 10 mM Na(+)-containing solution, nifedipine application before a +10 mV test pulse blocked 62% of the rapid initial phase of the indo-1 transient. Calibration curves of indo-1 for intracellular Ca2+ (using a KD of indo-1 for Ca2+ of either 250 or 850 nM, the reported range) indicated that between 67 and 76% of the Ca2+i transient was inhibited by nifedipine. Thus, in cells dialysed with 10 mM Na+ and depolarized to +10 mV, and in the absence of ICa,L, this suggests that another trigger mechanism for SR release is able to trigger between 33 and 24% of the Ca2+i transient. 5. For a given dialysing Na+ concentration, the fraction of indo-1 transient which was inhibited by nifedipine decreased as test potential became more positive. In cells dialysed with 10 mM Na+ and pulsed to +110 mV, 24% of the rapid phase of the indo-1 transient was inhibited by nifedipine (equivalent to between 27 and 37% of the Ca2+i transient). 6. For a given test potential, the fraction of the indo-1 transient which was inhibited by nifedipine decreased as dialysing Na+ concentration increased. In cells dialysed with Na(+)-free solution and pulsed to +10 mV, 84% of the indo-1 transient was inhibited by nifedipine (equivalent to between 88 and 91% of the Ca2+i transient). In contrast, in cells dialysed with 20 mM Na+ and pulsed to +10 mV, 41% of the indo-1 transient was inhibited by nifedipine (equivalent to between 47 and 57% of the Ca2+i transient). 7. Dialysing cells with different Na+ concentrations could lead to a different SR Ca2+ content. We therefore manipulated the conditioning train before each test pulse to change the extent of SR loading. For each dialysing Na+ concentration, we found no change in the degree to which nifedipine blocked the indo-1 transient when SR content was either increased or decreased. 8. The results support the idea that both ICa, L and a second mechanism are able to trigger SR release and the resulting Ca2+i transient. When ICa, L was blocked with nifedipine, the fraction of Ca2+i transient which remained increased with more positive test potential and higher internal Na+. This is consistent with the hypothesis that the second SR trigger mechanism is Ca2+ entry via reverse Na(+)-Ca2+ exchange, elicited by a step change in membrane potential.
Collapse
Affiliation(s)
- A J Levi
- Department of Physiology, School of Medical Sciences, University of Bristol, UK.
| | | | | | | |
Collapse
|
28
|
Tinker A, Sutko JL, Ruest L, Deslongchamps P, Welch W, Airey JA, Gerzon K, Bidasee KR, Besch HR, Williams AJ. Electrophysiological effects of ryanodine derivatives on the sheep cardiac sarcoplasmic reticulum calcium-release channel. Biophys J 1996; 70:2110-9. [PMID: 9172735 PMCID: PMC1225186 DOI: 10.1016/s0006-3495(96)79777-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We have examined the effects of a number of derivatives of ryanodine on K+ conduction in the Ca2+ release channel purified from sheep cardiac sarcoplasmic reticulum (SR). In a fashion comparable to that of ryanodine, the addition of nanomolar to micromolar quantities to the cytoplasmic face (the exact amount depending on the derivative) causes the channel to enter a state of reduced conductance that has a high open probability. However, the amplitude of that reduced conductance state varies between the different derivatives. In symmetrical 210 mM K+, ryanodine leads to a conductance state with an amplitude of 56.8 +/- 0.5% of control, ryanodol leads to a level of 69.4 +/- 0.6%, ester A ryanodine modifies to one of 61.5 +/- 1.4%, 9,21-dehydroryanodine to one of 58.3 +/- 0.3%, 9 beta,21beta-epoxyryanodine to one of 56.8 +/- 0.8%, 9-hydroxy-21-azidoryanodine to one of 56.3 +/- 0.4%, 10-pyrroleryanodol to one of 52.2 +/- 1.0%, 3-epiryanodine to one of 42.9 +/- 0.7%, CBZ glycyl ryanodine to one of 29.4 +/- 1.0%, 21-p-nitrobenzoyl-amino-9-hydroxyryanodine to one of 26.1 +/- 0.5%, beta-alanyl ryanodine to one of 14.3 +/- 0.5%, and guanidino-propionyl ryanodine to one of 5.8 +/- 0.1% (chord conductance at +60 mV, +/- SEM). For the majority of the derivatives the effect is irreversible within the lifetime of a single-channel experiment (up to 1 h). However, for four of the derivatives, typified by ryanodol, the effect is reversible, with dwell times in the substate lasting tens of seconds to minutes. The effect caused by ryanodol is dependent on transmembrane voltage, with modification more likely to occur and lasting longer at +60 than at -60 mV holding potential. The addition of concentrations of ryanodol insufficient to cause modification does not lead to an increase in single-channel open probability, such as has been reported for ryanodine. At concentrations of > or = 500 mu M, ryanodine after initial rapid modification of the channel leads to irreversible closure, generally within a minute. In contrast, comparable concentrations of beta-alanyl ryanodine do not cause such a phenomenon after modification, even after prolonged periods of recording (>5 min). The implications of these results for the site(s) of interaction with the channel protein and mechanism of the action of ryanodine are discussed. Changes in the structure of ryanodine can lead to specific changes in the electrophysiological consequences of the interaction of the alkaloid with the sheep cardiac SR Ca2+ release channel.
Collapse
Affiliation(s)
- A Tinker
- Cardiac Medicine, National Heart and Lung Institute, Imperial College, University of London, London, England
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Levi AJ, Mitcheson JS, Hancox JC. The effect of internal sodium and caesium on phasic contraction of patch-clamped rabbit ventricular myocytes. J Physiol 1996; 492 ( Pt 1):1-19. [PMID: 8730578 PMCID: PMC1158856 DOI: 10.1113/jphysiol.1996.sp021284] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
1. The voltage dependence of phasic contraction was assessed in rabbit ventricular myocytes. Phasic contraction at all potentials was abolished by exposure to ryanodine-thapsigargin, showing that it was due primarily to Ca2+ release from the sarcoplasmic reticulum (SR). Experiments were performed at 35 degrees C, cells were whole-cell patch clamped and contraction was measured optically as unloaded shortening. Cells were held at -40 mV to inactivate the Na+ current (INa) and T-type Ca2+ current. A standard cellular Ca2+ load was established by applying a train of conditioning pulses at 0.5 Hz before each test pulse. The effect of replacing K+ with Cs+ in the dialysing pipette solution, and the effect of altering dialysing [Na+] between 0 and 20 mM, was assessed on contraction. 2. Cells dialysed with a K(+)-based, Na(+)-free solution exhibited a 'bell-shaped' voltage dependence of the L-type Ca2+ channel current (ICa,L), with a maximum ICa,L at +10 mV. Replacing internal K+ with Cs+, or altering pipette [Na+], did not affect the voltage dependence of ICa,L. 3. The voltage dependence of phasic contraction in cells dialysed with a K(+)-based solution was modulated by pipette [Na+]. The voltage dependence of phasic contraction was bell-shaped with 0 Na+, became much loss bell-shaped with 10 mM Na+ and with 20 mM Na+ the phasic contraction elicited at +100 mV was 1.6-fold larger than that at +10 mV. 4. Replacing 80% of K+ with Cs+ in the pipette dialysis solution led to a significant reduction in contraction amplitude and a more rapid decline in contraction amplitude after beginning the dialysis of the cell. 5. Cells dialysed with a Cs(+)-based solution displayed a voltage dependence of phasic contraction which was more bell-shaped (i.e. more similar to that of ICa,L) than that obtained with the corresponding K(+)-based dialysis solution. The level of pipette [Na+] still modulated the voltage dependence of phasic contraction in cells dialysed with a Cs(+)-based solution. 6. Time-to-peak contraction (tpk) also displayed voltage dependence; it had a minimum value between 0 and +20 mV (the voltage range for maximum ICa,L), but increased at more negative and positive potentials. Alteration of tpk contraction is discussed in relation to the stochastic behaviour of L-type Ca2+ channels and SR Ca2+ release channels. 7. The shape of the voltage dependence of contraction in rabbit myocytes at 35 degrees C is modulated by dialysing [Na+] over the tested range, 0-20 mM. Modulation of voltage dependence of contraction by dialysing [Na+] is consistent with an influence of reverse Na(+)-Ca2+ exchange in triggering intracellular Ca2+ release, in addition to the trigger Ca2+ which enters via ICa,L. 8. The marked effect of dialysing Cs+ on contraction amplitude, and on the voltage dependence of phasic contraction, does not appear to have been reported previously. Internal dialysis with Cs+ is a commonly used technique for blocking interfering outward K+ currents, in order to measure ICa,L more selectively. The present study suggests that Cs+ might also interfere with processes involved in excitation-contraction coupling and indicates that it might be wise to exercise caution with the use of internal Cs+ in experiments investigating excitation-contraction coupling.
Collapse
Affiliation(s)
- A J Levi
- Department of Physiology, School of Medical Sciences, University of Bristol, UK
| | | | | |
Collapse
|
30
|
Tsushima RG, Kelly JE, Wasserstrom JA. Characteristics of cocaine block of purified cardiac sarcoplasmic reticulum calcium release channels. Biophys J 1996; 70:1263-74. [PMID: 8785282 PMCID: PMC1225052 DOI: 10.1016/s0006-3495(96)79683-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have examined the effects of cocaine on the SR Ca2+ release channel purified from canine cardiac muscle. Cocaine induced a flicker block of the channel from the cytoplasmic side, which resulted in an apparent reduction in the single-channel current amplitude without a marked reduction in the single-channel open probability. This block was evident only at positive holding potentials. Analysis of the block revealed that cocaine binds to a single site with an effective valence of 0.93 and an apparent dissociation constant at 0 mV (Kd(0)) of 38 mM. The kinetics of cocaine block were analyzed by amplitude distribution analysis and showed that the voltage and concentration dependence lay exclusively in the blocking reaction, whereas the unblocking reaction was independent of both voltage and concentration. Modification of the channel by ryanodine dramatically attenuated the voltage and concentration dependence of the on rates of cocaine block while diminishing the off rates to a lesser extent. In addition, ryanodine modification changed the effective valence of cocaine block to 0.52 and the Kd(0) to 110 mM, suggesting that modification of the channel results in an alteration in the binding site and its affinity for cocaine. These results suggest that cocaine block of the SR Ca2+ release channel is due to the binding at a single site within the channel pore and that modification of the channel by ryanodine leads to profound changes in the kinetics of cocaine block.
Collapse
Affiliation(s)
- R G Tsushima
- Department of Medicine (Cardiology), Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
31
|
Abstract
In this review, we summarized the results obtained mainly by flux measurements through Ca2+ channel in HSR vesicles. The Ca2+ channel has a large pore which passes not only divalent cations such as Ca2+, Mg2+, and Ba2+ and monovalent cations such as Na+, K+, and Cs+, but also large ions such as choline and tris. The permeation rates of choline and glucose through the Ca2+ channel were measured quantitatively by the light scattering method. The slow permeation of such molecules may reflect the structure of pores since the permeation process is the rate-limiting step for such large molecules. Neutral molecules such as glucose became permeable in the presence of submolar KCl, which suggests that pore size of the channel becomes larger in KCl. The apparent permeation rates of Ca2+ and Mg2+ obtained from the flux measurement were the same, although their single-channel conductances were different. This discrepancy was explained by the fact that flux measurements reflects the open rate of the channel. Thus, complementarity between the flux measurement and single-channel recording was demonstrated. From the effects of K+ on the action of regulators on Ca2+ channel, it was suggested that the Ca2+ channel has many binding sites for activators and inhibitors. There are two kinds of Ca2+ binding sites for activation and inhibition. Activation sites for Ca2+, caffeine, and ATP are different and inhibition sites for Ca2+ and procaine are different. The binding sites for ruthenium red and Mg2+ are the same as the activation and/or inhibition sites for Ca2+. Ryanodine-treated Ca2+ channel became permeable to glucose even in the absence of KCl. The conformational state of the channel opened by ryanodine is different from that opened by Ca2+, caffeine, and ATP. The maximal flux rates of choline and glucose induced by ryanodine were smaller than those attained by caffeine and ATP. This result is consistent with the observation obtained by single-channel recording; the maximal value of single-channel conductance after ryanodine treatment becomes 40-50% of the value before the treatment. It is likely that the radius of the pore opened by ryanodine is smaller than that opened by Ca2+, caffeine, or ATP. The flexibility of the channel may be decreased in the open locked state induced by ryanodine. The Ca2+ response to open the channel by micromolar Ca2+ was lost when calsequestrin was released from the vesicles. It is possible that calsequestrin acts as an endogenous regulator of Ca2+ channel through triadin in excitation-contraction coupling.
Collapse
Affiliation(s)
- M Kasai
- Department of Biophysical Engineering, Faculty of Engineering Science, Osaka University, Japan
| | | |
Collapse
|
32
|
Schiefer A, Meissner G, Isenberg G. Ca2+ activation and Ca2+ inactivation of canine reconstituted cardiac sarcoplasmic reticulum Ca(2+)-release channels. J Physiol 1995; 489 ( Pt 2):337-48. [PMID: 8847630 PMCID: PMC1156762 DOI: 10.1113/jphysiol.1995.sp021055] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
1. Calcium-release channels (ryanodine receptors) of canine cardiac sarcoplasmic reticulum (SR) were incorporated into lipid bilayer membranes at the tip of a patch pipette. Using symmetrical 150 mM KCl solutions, [Ca2+] > 0.3 microM activated single channels of 627 pS conductance. The kinetics of Ca(2+)-mediated channel activation, deactivation and inactivation were studied by stepwise changes in pCa (-log[Ca2+]) and analysis of current means. 2. Steps of [Ca2+] activated the channel open probability (Po) along a time course which could be fitted by a single exponential. The activation time constant was dependent on [Ca2+], which decreased from 4.9 ms at pCa 6.5 to 0.2 ms at pCa 3. Subsequent rapid reduction in [Ca2+] decreased Po along a mono-exponential deactivation time course, the time constant of which was independent of the [Ca2+] during the preceding activation period. Further analysis yielded the rate constants kon of 2 x 10(8) (M s)-1 and koff of 2 x 10(2) s-1, an apparent dissociation constant (KD) of 1 microM, and a Hill coefficient of 1.05. 3. The open probability increased with [Ca2+], reaching a peak at about pCa 5.5. At pCa < or = 5.5, Po decreased time dependently, the time constants decreasing along with [Ca2+] from 1 s at 3 microM to 0.2 s at 1 mM. During the 0.5 s period at 3 microM Ca2+, Po fell by 13% due to an extension of the closed times. At 1 mM Ca2+, Po 'inactivated' by 72%, which was due mostly to long closures. These differences suggest that the Ca(2+)-mediated decay of Po was dependent on Ca2+ binding to an intermediate (KD, 3 microM) and a low affinity site (KD, 360 microM). On the return of pCa from 3 to > 8, the channels briefly re-opened. 4. A 'refractory' behaviour of the channel was not observed for 20 ms steps between < 10 nM and < 10 microM [Ca2+] (25 Hz). For steps between 10 nM and 1 mM, however, such behaviour was marked by infrequent and irregular channel openings. 5. The results are described by a three Ca2+ binding site model and compared with the literature.
Collapse
Affiliation(s)
- A Schiefer
- Department of Physiology, University of Cologne, Germany
| | | | | |
Collapse
|
33
|
Shoshan-Barmatz V, Hadad-Halfon N, Ostersetzer O. Cross-linking of the ryanodine receptor/Ca2+ release channel from skeletal muscle. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1237:151-61. [PMID: 7632708 DOI: 10.1016/0005-2736(95)00084-g] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The relationship between the tetrameric organization of the ryanodine receptor (RyR) and its activity in binding of ryanodine was approached through cross-linking studies using several bifunctional reagents, differing in their linear dimensions and flexibility, as well as in the reactivity of the active groups. Cross-linking with: 1,5-difluoro-2,4-dinitrobenzene (DFDNB); di(fluoro-3-nitrophenyl)sulfone (DFNPS), 1-ethyl-3-(3-dimethylamino)propyl)carbodiimide (EDC); dimethyl suberimidate (DMS); ethylene glycol bis(succinimidylsuccinate) (EGS); and glutaraldehyde resulted in the disappearance of the, 470 kDa, RyR monomer protein band with concomitant appearance of additional bands of molecular masses higher than the monomer. At the relatively low concentrations of the reagents and the conditions used, RyR is the only cross-linked protein of SR membranes. The 'new' protein bands cross-react with antibodies against the RyR and correspond to dimers and tetramers of the RyR subunits while trimers were not detectable. DFDNB and DFNPS produced also a 560 kDa protein band which probably represents an intramolecular cross-linked monomer. The SDS-electrophoretic patterns of the cross-linked purified RyR resemble those of the membrane-bound receptor. Ryanodine binding to the high-affinity site was inhibited by modification of SR membranes with DFDNB and DFNPS, but not with DMS, EDC, EGS and glutaraldehyde, although RyR was completely cross-linked. The inhibition by DFDNB and DFNPS is due to modification of a specific lysyl residue which is also involved in the control of Ca2+ release. On the other hand, cross linking of the RyR with glutaraldehyde or EGS resulted in inhibition of ryanodine binding to the low-affinity, but not to the high-affinity binding sites. Thus, the cross-linking of two or more sites in each monomer (which lead to fixation of dimers or tetramers) did not prevent the conformational changes involved in the binding and occlusion of ryanodine at the high-affinity site, but inhibited its binding to the low-affinity sites.
Collapse
Affiliation(s)
- V Shoshan-Barmatz
- Department of Life Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | | |
Collapse
|
34
|
Tinker A, Williams AJ. Measuring the length of the pore of the sheep cardiac sarcoplasmic reticulum calcium-release channel using related trimethylammonium ions as molecular calipers. Biophys J 1995; 68:111-20. [PMID: 7536054 PMCID: PMC1281667 DOI: 10.1016/s0006-3495(95)80165-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
After incorporation of purified sheep cardiac Ca(2+)-release channels into planar phospholipid bilayers, we have investigated the blocking effects of a series of monovalent (CH3-(CH2)n-1-N+(CH3)3) and divalent ((CH3)3N(+)-(CH2)n-N+(CH3)3) trimethylammonium derivatives under voltage clamp conditions. All the compounds tested produce voltage-dependent block from the cytoplasmic face of the channel. With divalent (Qn) derivatives the effective valence of block decreases with increasing chain length, reaching a plateau with a chain length of n > or = 7. No decline in effective valence is observed with the monovalent (Un) derivatives. A plausible interpretation of this phenomena suggests that for the 90% of the voltage drop measured, the increase in length following the addition of a CH2 in the chain spans 12.7% of the electrical field. Extrapolating this distance to include the remaining 10% suggests that the applied holding potential falls over a total distance of 10.4 A. In addition, at high positive holding potentials there is evidence for permeation of the trimethylammonium ions and a valency specific relief of block.
Collapse
Affiliation(s)
- A Tinker
- Department of Cardiac Medicine, National Heart and Lung Institute, University of London, United Kingdom
| | | |
Collapse
|
35
|
Percival AL, Williams AJ, Kenyon JL, Grinsell MM, Airey JA, Sutko JL. Chicken skeletal muscle ryanodine receptor isoforms: ion channel properties. Biophys J 1994; 67:1834-50. [PMID: 7532019 PMCID: PMC1225557 DOI: 10.1016/s0006-3495(94)80665-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
To define the roles of the alpha- and beta-ryanodine receptor (RyR) (sarcoplasmic reticulum Ca2+ release channel) isoforms expressed in chicken skeletal muscles, we investigated the ion channel properties of these proteins in lipid bilayers. alpha- and beta RyRs embody Ca2+ channels with similar conductances (792, 453, and 118 pS for K+, Cs+ and Ca2+) and selectivities (PCa2+/PK+ = 7.4), but the two channels have different gating properties. alpha RyR channels switch between two gating modes, which differ in the extent they are activated by Ca2+ and ATP, and inactivated by Ca2+. Either mode can be assumed in a spontaneous and stable manner. In a low activity mode, alpha RyR channels exhibit brief openings (tau o = 0.14 ms) and are minimally activated by Ca2+ in the absence of ATP. In a high activity mode, openings are longer (tau o1-3 = 0.17, 0.51, and 1.27 ms), and the channels are activated by Ca2+ in the absence of ATP and are in general less sensitive to the inactivating effects of Ca2+. beta RyR channel openings are longer (tau 01-3 = 0.34, 1.56, and 3.31 ms) than those of alpha RyR channels in either mode. beta RyR channels are activated to a greater relative extent by Ca2+ than ATP and are inactivated by millimolar Ca2+ in the absence, but not the presence, of ATP. Both alpha- and beta RyR channels are activated by caffeine, inhibited by Mg2+ and ruthenium red, inactivated by voltage (cytoplasmic side positive), and modified to a long-lived substate by ryanodine, but only alpha RyR channels are activated by perchlorate anions. The differences in gating and responses to channel modifiers may give the alpha- and beta RyRs distinct roles in muscle activation.
Collapse
Affiliation(s)
- A L Percival
- Department of Pharmacology, University of Nevada School of Medicine, Reno 89557
| | | | | | | | | | | |
Collapse
|
36
|
Single-channel inositol 1,4,5-trisphosphate receptor currents revealed by patch clamp of isolated Xenopus oocyte nuclei. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(18)43887-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
37
|
Ahern GP, Junankar PR, Dulhunty AF. Single channel activity of the ryanodine receptor calcium release channel is modulated by FK-506. FEBS Lett 1994; 352:369-74. [PMID: 7523191 DOI: 10.1016/0014-5793(94)01001-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The immunosuppressant drug FK-506 (3-20 microM) increased the open probability of ryanodine receptor calcium release channels, formed by incorporation of terminal cisternae vesicles from rabbit skeletal muscle into lipid bilayers, with cis (cytoplasmic) calcium concentrations between 10(-7) M and 10(-3) M. FK-506 increased mean current and channel open time and induced long sojourns at subconductance levels that were between 28% and 38% of the maximum conductance and were distinct from the ryanodine-induced subconductance level at about 45% of the maximum conductance. FK-506 relieved the Ca2+ inactivation of the ryanodine receptor seen at 10(-3) M Ca2+. The results are consistent with FK-506 removal of FK-506 binding protein from the ryanodine receptor.
Collapse
Affiliation(s)
- G P Ahern
- Division of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra City
| | | | | |
Collapse
|
38
|
Sitsapesan R, Williams AJ. Gating of the native and purified cardiac SR Ca(2+)-release channel with monovalent cations as permeant species. Biophys J 1994; 67:1484-94. [PMID: 7819484 PMCID: PMC1225511 DOI: 10.1016/s0006-3495(94)80622-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The primary aim of this study was to characterize the steady-state gating of the native and the purified cardiac sarcoplasmic reticulum Ca(2+)-release channel using monovalent cations (K+ in the purified, Cs+ in the native) rather than Ca2+ as the permeant ions. The improved resolution of the single-channel events under these conditions has provided a more detailed and accurate description of channel gating than was previously possible. Micromolar cytosolic Ca2+ activates the channel but in the absence of other activating ligands cannot fully open the channel. The relationship between the open probability (Po) and cytosolic free [Ca2+] in both native and purified channels indicates the binding of at least three Ca2+ ions for maximal activation. Lifetime analysis indicates a minimum of three open and five closed states for channels activated solely by Ca2+ and demonstrates that the primary mechanism for the increase in Po is an increase in the frequency of channel opening. Burst analysis also indicates that Ca2+ activates the channel by binding to closed states of the channel to increase the frequency of channel opening. Correlations between successive lifetimes suggest the existence of at least two pathways between the open and closed states. At a given activating [Ca2+], the Po is lower at negative than at positive holding potentials; however, we find no change in the mechanisms of Ca2+ activation at different voltages. Po measurements and lifetime analysis indicate that the gating of the purified channel when activated by Ca2+ is indistinguishable from that of the native channel and indicate that the channels are not modified by the purification procedure.
Collapse
Affiliation(s)
- R Sitsapesan
- Department of Cardiac Medicine, National Heart and Lung Institute, University of London, United Kingdom
| | | |
Collapse
|
39
|
Mygland A, Aarli JA, Matre R, Gilhus NE. Ryanodine receptor antibodies related to severity of thymoma associated myasthenia gravis. J Neurol Neurosurg Psychiatry 1994; 57:843-6. [PMID: 8021674 PMCID: PMC1073029 DOI: 10.1136/jnnp.57.7.843] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ryanodine receptor (RyR) antibodies are detected in about 50% of patients with myasthenia gravis who have a thymoma. The RyR is a calcium release channel involved in the mechanism of excitation-contraction coupling in striated muscle. In this study the severity of myasthenia gravis assessed by a five point disability score was compared between 12 patients with myasthenia gravis, a thymoma, and RyR antibodies and 10 patients with myasthenia gravis and a thymoma but without such antibodies. Symptoms of myasthenia gravis were significantly more severe in patients with RyR antibodies. The mean (SD) disability scores were 3.7(0.5) in patients with antibodies and 2.7 (0.9) in those without at peak of illness, (p = 0.01) and 3.4(1.4) v 1.6(0.7) at the end of an average observation period of five years (p = 0.002). The number of deaths due to myasthenia gravis was five of 12 RyR antibody positive patients, and none of 10 RyR antibody negative patients (p = 0.04). RyR antibody levels correlated positively with severity of myasthenia gravis. The presence of circulating RyR antibodies seems to be associated with a severe form of thymoma associated myasthenia gravis.
Collapse
Affiliation(s)
- A Mygland
- Department of Neurology, Gade Institute, University of Bergen, Norway
| | | | | | | |
Collapse
|
40
|
Brillantes AB, Ondrias K, Scott A, Kobrinsky E, Ondriasová E, Moschella MC, Jayaraman T, Landers M, Ehrlich BE, Marks AR. Stabilization of calcium release channel (ryanodine receptor) function by FK506-binding protein. Cell 1994; 77:513-23. [PMID: 7514503 DOI: 10.1016/0092-8674(94)90214-3] [Citation(s) in RCA: 587] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
FK506-binding protein (FKBP12) was originally identified as the cytosolic receptor for the immunosuppressant drugs FK506 and rapamycin. The cellular function of FKBP12, a ubiquitously expressed 12,000-dalton proline isomerase, has been unknown. FKBP12 copurifies with the 565,000-dalton ryanodine receptor (RyR), four of which form intracellular Ca2+ release channels of the sarcoplasmic and endoplasmic reticula. By coexpressing the RyR and FKBP12 in insect cells, we have demonstrated that FKBP12 modulates channel gating by increasing channels with full conductance levels (by > 400%), decreasing open probability after caffeine activation (from 0.63 +/- 0.09 to 0.04 +/- 0.02), and increasing mean open time (from 4.4 +/- 0.6 ms to 75 +/- 41 ms). FK506 or rapamycin, inhibitors of FKBP12 isomerase activity, reverse these stabilizing effects. These results provide the first natural cellular function for FKBP12, and establish that the functional Ca2+ release channel complex includes FKBP12.
Collapse
Affiliation(s)
- A B Brillantes
- Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Spät A, Rohács T, Hunyady L. Plasmalemmal dihydropyridine receptors modify the function of subplasmalemmal inositol 1,4,5-trisphosphate receptors: a hypothesis. Cell Calcium 1994; 15:431-7. [PMID: 8033201 DOI: 10.1016/0143-4160(94)90018-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Experimental observations on rat glomerulosa cells inspired a model which postulates that plasmalemmal dihydropyridine receptors are in juxtaposition and interaction with inositol 1,4,5-trisphosphate receptors in subplasmalemmal calciosomes. Activation of dihydropyridine receptors promotes the Ca2+ releasing effect of inositol 1,4,5-trisphosphate. The most important observations compatible with the model are the following: (1) angiotensin II does not influence Ca2+ influx during the peak phase of Ca2+ signal; (2) dihydropyridine drugs modify the initial peak of the Ca2+ signal induced by angiotensin II; (3) inhibitors of the dihydropyridine receptor reduce the initial Ca2+ signal also in the presence of 5 mM Ni2+, an inhibitor of voltage dependent Ca2+ influx; and (4) changes in extracellular K+ concentration within the physiological range also modify the cytoplasmic Ca2+ response to angiotensin II.
Collapse
Affiliation(s)
- A Spät
- Department of Physiology, Semmelweis University of Medicine, Budapest, Hungary
| | | | | |
Collapse
|
42
|
Abstract
Recent findings on the ryanodine receptor of vertebrates, a Ca-release channel protein for the caffeine- and ryanodine-sensitive Ca pools, are reviewed in this article. Three distinct genes, i.e., ryr1, ryr2, and ryr3, express different isoforms in specific locations: Ryr1 in skeletal muscle and Purkinje cells of cerebellum; Ryr2 in cardiac muscle and brain, especially cerebellum; Ryr3 in skeletal muscle of nonmammalian vertebrates, the corpus striatum, and limbic cortex of brain, smooth muscles, and the other cells in vertebrates. While only one isoform (Ryr1) is expressed in mammalian skeletal muscles, two isoforms (alpha- and beta-isoforms expressed by ryr1 and ryr3, respectively) are found in nonmammalian vertebrate skeletal muscles. Although the coexistence of two isoforms may merely be related to differentiation and specialization, the biological significance remains to be clarified. Ryanodine receptors in vertebrate skeletal muscles are believed to mediate two different modes of Ca release: Ca(2+)-induced Ca release and action potential-induced Ca release. All results obtained so far with any isoform of ryanodine receptor are related to Ca(2+)-induced Ca release and show very similar characteristics. Ca(2+)-induced Ca release, however, cannot be the underlying mechanism of Ca release on skeletal muscle activation. Susceptibility of the ryanodine receptor's ryanodine-binding activity to modification by physical factors, such as osmolality of the medium, might be related to action potential-induced Ca release. A hypothesis of molecular interaction in view of the plunger model of action potential-induced Ca release is discussed, suggesting that the model could be compatible with Ryr1 and Ryr3, but incompatible with Ryr2. The functional relevance of ryanodine receptor isoforms, especially Ryr3, in brain also remains to be clarified. Among ryr1 gene-related diseases, malignant hyperthermia was the first to be identified; however, there is still the possibility of involvement of the other genes. Central core disease has been added to the list recently. A molecular approach for the diagnosis and treatment of diseases is now in progress.
Collapse
Affiliation(s)
- Y Ogawa
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo
| |
Collapse
|
43
|
Schaffer SW, Ballard C, Azuma J. Mechanisms underlying physiological and pharmacological actions of taurine on myocardial calcium transport. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1994; 359:171-80. [PMID: 7887258 DOI: 10.1007/978-1-4899-1471-2_18] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- S W Schaffer
- Department of Pharmacology, School of Medicine, University of South Alabama, Mobile
| | | | | |
Collapse
|
44
|
Hohenegger M, Suko J. Phosphorylation of the purified cardiac ryanodine receptor by exogenous and endogenous protein kinases. Biochem J 1993; 296 ( Pt 2):303-8. [PMID: 8257417 PMCID: PMC1137694 DOI: 10.1042/bj2960303] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The ryanodine receptor is the main Ca(2+)-release structure in skeletal and cardiac sarcoplasmic reticulum. In both tissues, phosphorylation of the ryanodine receptor has been proposed to be involved in the regulation of Ca2+ release. In the present study, we have examined the ability of the purified cardiac ryanodine receptor to serve as a substrate for phosphorylation by exogenously added catalytic subunit of the cyclic AMP (cAMP)-dependent protein kinase (PK-A), cyclic GMP (cGMP)-dependent protein kinase (PK-G), or calmodulin-dependent protein kinase (PK-CaM). A large amount of phosphate incorporation was observed for PK-CaM (938 +/- 48 pmol of Pi/mg of purified channel protein), whereas the level of phosphorylation was considerably lower with PK-A or PK-G (345 +/- 139 and 96 +/- 6 pmol/mg respectively). In addition, endogenous PK-CaM activity co-migrates with the ryanodine receptor through several steps of purification, suggesting a strong association of the two proteins. This endogenous PK-CaM activity is abolished by a PK-CaM-specific synthetic peptide inhibitor. Endogenous cAMP- and cGMP-dependent phosphorylation was not observed in the purified ryanodine-receptor preparation. Taken together, these observations imply that PK-CaM is the physiologically relevant protein kinase, capable of phosphorylating the channel protein to a minimum stoichiometry of 2 mol of Pi per mol of tetramer.
Collapse
Affiliation(s)
- M Hohenegger
- Institute of Pharmacology, University of Vienna, Austria
| | | |
Collapse
|
45
|
Leddy JJ, Murphy BJ, Doucet JP, Pratt C, Tuana BS. A 60 kDa polypeptide of skeletal-muscle sarcoplasmic reticulum is a calmodulin-dependent protein kinase that associates with and phosphorylates several membrane proteins. Biochem J 1993; 295 ( Pt 3):849-56. [PMID: 8240301 PMCID: PMC1134639 DOI: 10.1042/bj2950849] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Activation of a calmodulin (CaM)-dependent protein kinase associated with rabbit skeletal-muscle sarcoplasmic reticulum (SR) results in the phosphorylation of polypeptides of 450, 360, 165, 105, 89, 60, 34 and 20 kDa. Radioligand-binding studies indicated that a membrane-bound 60 kDa polypeptide contained both CaM- and ATP-binding domains. Under renaturing conditions on nitrocellulose blots, the 60 kDa polypeptide of the membrane exhibited CaM-dependent autophosphorylation activity, suggesting that it was the CaM-dependent protein kinase of SR. Ca2+/CaM-independent autophosphorylation of polypeptides of 62 and 45 kDa was found to occur in the light SR, whereas the Ca2+/CaM-dependent autophosphorylation activity was enriched in the heavy SR. Both these kinase activities were absent from transverse tubules, although these membranes were enriched in CaM-binding polypeptides of 160, 100 and 80 kDa. In the absence of Ca2+, CaM bound to a 33 kDa polypeptide of the membrane. The purified ryanodine receptor was not phosphorylated by the purified CaM kinase, although it was a substrate for protein kinase C. Affinity-purified antibodies to brain CaM kinase II cross-reacted with the 60 kDa polypeptide in Western blots and immunoprecipitated the 60 kDa polypeptide, along with the 360, 105, 89, 34 and 20 kDa phosphoproteins, from Nonidet-P-40-solubilized SR membranes. Antibodies raised against the 60 kDa kinase polypeptide did not cross-react with the other phosphoproteins, suggesting that these polypeptides were distinct and unrelated. Subcellular distribution of the 60 kDa kinase indicated the specific association of the polypeptide with the junctional-face membrane of SR. The CaM-dependent incorporation of 32P into various membrane proteins was inhibited by the CaM kinase II fragment (290-309), with an IC50 value of 2 nM for the inhibition of incorporation into the 60 kDa kinase polypeptide. Recent studies [Wang and Best (1992) Nature (London) 359, 739-741] have shown that a CaM kinase activity intrinsic to the membrane can inactivate the Ca(2+)-release channel of skeletal muscle SR. Since our results demonstrate that the 60 kDa polypeptide of SR is a CaM-dependent protein kinase, we suggest that this kinase, through its associations, may be responsible for gating the Ca(2+)-release channel.
Collapse
Affiliation(s)
- J J Leddy
- Department of Pharmacology, University of Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
46
|
Tinker A, Williams AJ. Charged local anesthetics block ionic conduction in the sheep cardiac sarcoplasmic reticulum calcium release channel. Biophys J 1993; 65:852-64. [PMID: 8218909 PMCID: PMC1225786 DOI: 10.1016/s0006-3495(93)81104-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We have examined the effect of the charged local anesthetics QX314, QX222, and Procaine on monovalent cation conduction in the Ca2+ release channel of the sheep cardiac sarcoplasmic reticulum. All three blockers only affect cation conductance when present at the cytoplasmic face of the channel. QX222 and Procaine act as voltage-dependent blockers. With 500 Hz filtering, this is manifest as a relatively smooth reduction in single-channel current amplitude most prominent at positive holding potentials. Quantitative analysis gives an effective valence of approximately 0.9 for both ions and Kb(0)s of 9.2 and 15.8 mM for QX222 and Procaine, respectively. Analysis of the concentration dependence of block suggests that QX222 is binding to a single site with a Km of 491 microM at a holding potential of 60 mV. The use of amplitude distribution analysis, with the data filtered at 1 to 2 kHz, reveals that the voltage and concentration dependence of QX222 block occurs largely because of changes in the blocker on rate. The addition of QX314 has a different effect, leading to the production of a substate with an amplitude of approximately one-third that of the control. The substate's occurrence is dependent on holding potential and QX314 concentration. Quantitative analysis reveals that the effect is highly voltage dependent, with a valence of approximately 1.5 caused by approximately equal changes in the on and off rates. Kinetic analysis of the concentration dependence of the substate occurrence reveals positive cooperativity with at least two QX314s binding to the conduction pathway, and this is largely accounted for by changes in the on rate. A paradoxical increase in the off rate at high positive holding potentials and with increasing QX314 concentration at 80 mV suggests the existence of a further QX314-dependent reaction that is both voltage and concentration dependent. The substate block is interpreted physically as a form of partial occlusion in the vestibule of the conduction pathway giving a reduction in single-channel current by electrostatic means.
Collapse
Affiliation(s)
- A Tinker
- Department of Cardiac Medicine, National Heart and Lung Institute, University of London, England
| | | |
Collapse
|
47
|
McGarry SJ, Williams AJ. Digoxin activates sarcoplasmic reticulum Ca(2+)-release channels: a possible role in cardiac inotropy. Br J Pharmacol 1993; 108:1043-50. [PMID: 8387382 PMCID: PMC1908139 DOI: 10.1111/j.1476-5381.1993.tb13503.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
1. The effect of digoxin on rapid 45Ca2+ efflux from cardiac and skeletal sarcoplasmic reticulum (SR) vesicles was investigated. Additionally the interaction of digoxin with single cardiac and skeletal muscle SR Ca(2+)-release channels incorporated into planar phospholipid bilayers and held under voltage clamp was determined. 2. Digoxin (1 nM) increased the initial rate and amount of Ca(2+)-induced release of 45Ca2+ from cardiac SR vesicles, passively loaded with 45CaCl2, at an extravesicular [Ca2+] of 0.1 microM. The efflux in the presence and absence of digoxin was inhibited at pM extravesicular Ca2+ and blocked by 5 mM Mg2+. 3. To elucidate the mechanism of action of digoxin, single-channel recording was used. Digoxin (1-20 nM) increased single-channel open probability (Po) when added to the cytosolic but not the luminal face of the cardiac channel in the presence of sub-maximally activating Ca2+ (0.1 microM-10 microM) with an EC50 of 0.91 nM at 10 microM Ca2+. The mechanisms underlying the action of digoxin appear to be concentration-dependent. The activation observed at 1 nM digoxin appears to be consistent with the sensitization of the channel to the effects of Ca2+. At higher concentrations the drug appears to interact synergistically with Ca2+ to produce values of Po considerably greater than those seen with Ca2+ as the sole activating ligand. 4. Digoxin had no effect on single-channel conductance or the Ca2+/Tris permeability ratio. In channels activated by digoxin the Po was decreased by Mg2+. Single-channels were characteristically modified to along lasting open, but reduced, conductance state when 100 nM ryanodine was added to the cytosolic side of the channel.5. Activation of the cardiac SR Ca2+-release channel was observed with similar concentrations of digitoxin, however, higher concentrations of ouabain were required to increase PO. In contrast, a steroid which is not positively inotropic, chlormadinone acetate, had no effect on either cardiac or skeletal SR Ca2+-release channel activity.6. At concentrations up to 1 microM, digoxin had no effect on Ca2+-induced 45Ca2+ efflux from skeletal muscle SR vesicles nor did it affect skeletal SR Ca2+-release channel Po, reflecting a difference between the cardiac and skeletal isoforms of the Ca2+-release channel.7. Since activation of the cardiac SR Ca2+-release channel occurs within the range of concentrations of digoxin encountered therapeutically, it is possible that activation of this channel contributes to the positive inotropic effect observed with this drug. Further, activation of the channel by higher concentrations of digoxin may contribute to the toxic effects seen clinically.
Collapse
Affiliation(s)
- S J McGarry
- Department of Cardiac Medicine, National Heart and Lung Institute, University of London
| | | |
Collapse
|
48
|
Csernoch L, Jacquemond V, Schneider MF. Microinjection of strong calcium buffers suppresses the peak of calcium release during depolarization in frog skeletal muscle fibers. J Gen Physiol 1993; 101:297-333. [PMID: 8384243 PMCID: PMC2216760 DOI: 10.1085/jgp.101.2.297] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The effects of high intracellular concentrations of various calcium buffers on the myoplasmic calcium transient and on the rate of release of calcium (Rrel) from the sarcoplasmic reticulum (SR) were studied in voltage-clamped frog skeletal muscle fibers. The changes in intracellular calcium concentration (delta[Ca2+]) for 200-ms pulses to 0-20 mV were recorded before and after the injection of the calcium buffer and the underlying Rrel was calculated. If the buffer concentration after the injection was high, the initial rate of rise of the calcium transient was slower after injection than before and was followed by a slow increase of [Ca2+] that resembled a ramp. The increase in myoplasmic [Mg2+] that accompanies the calcium transient in control was suppressed after the injection and a slight decrease was observed instead. After the injection the buffer concentration in the voltage-clamped segment of the fiber decreased as the buffer diffused away toward the open ends. The calculated apparent diffusion coefficient for fura-2 (Dapp = 0.40 +/- 0.03 x 10(-6) cm2/s, mean +/- SEM, n = 6) suggests that approximately 65-70% of the indicator was bound to relatively immobile intracellular constituents. As the concentration of the injected buffer decreased, the above effects were reversed. The changes in delta[Ca2+] were underlined by characteristic modification of Rrel. The early peak component was suppressed or completely eliminated; thus, Rrel rose monotonically to a maintained steady level if corrected for depletion. If Rrel was expressed as percentage of SR calcium content, the steady level after injection did not differ significantly from that before. Control injections of anisidine, to the concentration that eliminated the peak of Rrel when high affinity buffers were used, had only a minor effect on Rrel, the peak was suppressed by 26 +/- 5% (mean +/- SE, n = 6), and the steady level remained unchanged. Thus, the peak component of Rrel is dependent on a rise in myoplasmic [Ca2+], consistent with calcium-induced calcium release, whereas the steady component of Rrel is independent of myoplasmic [Ca2+].
Collapse
Affiliation(s)
- L Csernoch
- Department of Biological Chemistry, University of Maryland, School of Medicine, Baltimore 21201
| | | | | |
Collapse
|