1
|
Campbell HM, Quick AP, Abu-Taha I, Chiang DY, Kramm CF, Word TA, Brandenburg S, Hulsurkar M, Alsina KM, Liu HB, Martin B, Uhlenkamp D, Moore OM, Lahiri SK, Corradini E, Kamler M, Heck AJR, Lehnart SE, Dobrev D, Wehrens XHT. Loss of SPEG Inhibitory Phosphorylation of Ryanodine Receptor Type-2 Promotes Atrial Fibrillation. Circulation 2020; 142:1159-1172. [PMID: 32683896 DOI: 10.1161/circulationaha.120.045791] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Enhanced diastolic calcium (Ca2+) release through ryanodine receptor type-2 (RyR2) has been implicated in atrial fibrillation (AF) promotion. Diastolic sarcoplasmic reticulum Ca2+ leak is caused by increased RyR2 phosphorylation by PKA (protein kinase A) or CaMKII (Ca2+/calmodulin-dependent kinase-II) phosphorylation, or less dephosphorylation by protein phosphatases. However, considerable controversy remains regarding the molecular mechanisms underlying altered RyR2 function in AF. We thus aimed to determine the role of SPEG (striated muscle preferentially expressed protein kinase), a novel regulator of RyR2 phosphorylation, in AF pathogenesis. METHODS Western blotting was performed with right atrial biopsies from patients with paroxysmal AF. SPEG atrial knockout mice were generated using adeno-associated virus 9. In mice, AF inducibility was determined using intracardiac programmed electric stimulation, and diastolic Ca2+ leak in atrial cardiomyocytes was assessed using confocal Ca2+ imaging. Phosphoproteomics studies and Western blotting were used to measure RyR2 phosphorylation. To test the effects of RyR2-S2367 phosphorylation, knockin mice with an inactivated S2367 phosphorylation site (S2367A) and a constitutively activated S2367 residue (S2367D) were generated by using CRISPR-Cas9. RESULTS Western blotting revealed decreased SPEG protein levels in atrial biopsies from patients with paroxysmal AF in comparison with patients in sinus rhythm. SPEG atrial-specific knockout mice exhibited increased susceptibility to pacing-induced AF by programmed electric stimulation and enhanced Ca2+ spark frequency in atrial cardiomyocytes with Ca2+ imaging, establishing a causal role for decreased SPEG in AF pathogenesis. Phosphoproteomics in hearts from SPEG cardiomyocyte knockout mice identified RyR2-S2367 as a novel kinase substrate of SPEG. Western blotting demonstrated that RyR2-S2367 phosphorylation was also decreased in patients with paroxysmal AF. RyR2-S2367A mice exhibited an increased susceptibility to pacing-induced AF, and aberrant atrial sarcoplasmic reticulum Ca2+ leak, as well. In contrast, RyR2-S2367D mice were resistant to pacing-induced AF. CONCLUSIONS Unlike other kinases (PKA, CaMKII) that increase RyR2 activity, SPEG phosphorylation reduces RyR2-mediated sarcoplasmic reticulum Ca2+ release. Reduced SPEG levels and RyR2-S2367 phosphorylation typified patients with paroxysmal AF. Studies in S2367 knockin mouse models showed a causal relationship between reduced S2367 phosphorylation and AF susceptibility. Thus, modulating SPEG activity and phosphorylation levels of the novel S2367 site on RyR2 may represent a novel target for AF treatment.
Collapse
Affiliation(s)
- Hannah M Campbell
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Ann P Quick
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Issam Abu-Taha
- Institute of Pharmacology (I.A.-T., D.D.), University Duisburg-Essen, Germany
| | - David Y Chiang
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Medicine (Cardiovascular Division), Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.Y.C.)
| | - Carlos F Kramm
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Tarah A Word
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Sören Brandenburg
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Germany (S.B., D.U., S.E.L.)
| | - Mohit Hulsurkar
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Katherina M Alsina
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Hui-Bin Liu
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Institute of Clinical Pharmacy, the Second Affiliated Hospital of Harbin Medical University, China (H.-B.L.)
| | - Brian Martin
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Dennis Uhlenkamp
- Institute of Pharmacology (I.A.-T., D.D.), University Duisburg-Essen, Germany.,Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Germany (S.B., D.U., S.E.L.)
| | - Oliver M Moore
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Neuroscience (O.M.M., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Satadru K Lahiri
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX
| | - Eleonora Corradini
- Biomolecular Mass Spectrometry & Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands (E.C., A.J.R.H.)
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery Huttrop (M.K.), University Duisburg-Essen, Germany
| | - Albert J R Heck
- Biomolecular Mass Spectrometry & Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, The Netherlands (E.C., A.J.R.H.)
| | - Stephan E Lehnart
- Heart Research Center Göttingen, Department of Cardiology & Pneumology, University Medical Center Göttingen, Germany (S.B., D.U., S.E.L.)
| | | | - Xander H T Wehrens
- Cardiovascular Research Institute (H.M.C., A.P.Q., D.Y.C., C.F.K., T.A.W., M.H., K.MA., H.-B.L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology & Biophysics (H.M.C., A.P.Q., D.Y.C., F.K., T.A.W., M.H., K.M.A., H.-L., B.M., O.M.M., S.K.L., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Neuroscience (O.M.M., X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Medicine (Cardiology) (X.H.T.W.), Baylor College of Medicine, Houston, TX.,Department of Pediatrics (X.H.T.W.), Baylor College of Medicine, Houston, TX.,Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
2
|
Mahalingam M, Fessenden JD. Methods for labeling skeletal muscle ion channels site-specifically with fluorophores suitable for FRET-based structural analysis. Methods Enzymol 2015; 556:455-74. [PMID: 25857795 DOI: 10.1016/bs.mie.2014.11.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Skeletal muscle excitation-contraction coupling is triggered by the concerted action of two enormous Ca(2+) channel complexes, the dihydropyridine receptor and the type 1 ryanodine receptor. Recent advances in our understanding of the structure of these large Ca(2+) channels have been driven by fluorescence resonance energy transfer (FRET)-based analysis. A methodological challenge in conducting these FRET measurements is the ability to site-specifically label these huge ion channels with donor and acceptor fluorophores capable of undergoing energy transfer. In this chapter, we detail specific protocols for tagging large membrane proteins with these fluorescent probes using three orthogonal labeling methods: fluorescent protein fusions, biarsenical reagents directed to engineered tetracysteine tags, and Cy3/5 nitrilotriacetic acid conjugates that bind to poly-histidine tags.
Collapse
Affiliation(s)
- Mohana Mahalingam
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - James D Fessenden
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
3
|
Baker MR, Fan G, Serysheva II. Single-particle cryo-EM of the ryanodine receptor channel in an aqueous environment. Eur J Transl Myol 2015; 25:4803. [PMID: 25844145 PMCID: PMC4748972 DOI: 10.4081/ejtm.2015.4803] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/22/2014] [Accepted: 12/31/2014] [Indexed: 12/31/2022] Open
Abstract
Ryanodine receptors (RyRs) are tetrameric ligand-gated Ca2+ release channels that are responsible for the increase of cytosolic Ca2+ concentration leading to muscle contraction. Our current understanding of RyR channel gating and regulation is greatly limited due to the lack of a high-resolution structure of the channel protein. The enormous size and unwieldy shape of Ca2+ release channels make X-ray or NMR methods difficult to apply for high-resolution structural analysis of the full-length functional channel. Single-particle electron cryo-microscopy (cryo-EM) is one of the only effective techniques for the study of such a large integral membrane protein and its molecular interactions. Despite recent developments in cryo-EM technologies and break-through single-particle cryo-EM studies of ion channels, cryospecimen preparation, particularly the presence of detergent in the buffer, remains the main impediment to obtaining atomic-resolution structures of ion channels and a multitude of other integral membrane protein complexes. In this review we will discuss properties of several detergents that have been successfully utilized in cryo-EM studies of ion channels and the emergence of the detergent alternative amphipol to stabilize ion channels for structure-function characterization. Future structural studies of challenging specimen like ion channels are likely to be facilitated by cryo-EM amenable detergents or alternative surfactants.
Collapse
Affiliation(s)
| | | | - Irina I. Serysheva
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
4
|
Pinali C, Kitmitto A. Serial block face scanning electron microscopy for the study of cardiac muscle ultrastructure at nanoscale resolutions. J Mol Cell Cardiol 2014; 76:1-11. [PMID: 25149127 DOI: 10.1016/j.yjmcc.2014.08.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/31/2014] [Accepted: 08/12/2014] [Indexed: 12/28/2022]
Abstract
Electron microscopy techniques have made a significant contribution towards understanding muscle physiology since the 1950s. Subsequent advances in hardware and software have led to major breakthroughs in terms of image resolution as well as the ability to generate three-dimensional (3D) data essential for linking structure to function and dysfunction. In this methodological review we consider the application of a relatively new technique, serial block face scanning electron microscopy (SBF-SEM), for the study of cardiac muscle morphology. Employing SBF-SEM we have generated 3D data for cardiac myocytes within the myocardium with a voxel size of ~15 nm in the X-Y plane and 50 nm in the Z-direction. We describe how SBF-SEM can be used in conjunction with selective staining techniques to reveal the 3D cellular organisation and the relationship between the t-tubule (t-t) and sarcoplasmic reticulum (SR) networks. These methods describe how SBF-SEM can be used to provide qualitative data to investigate the organisation of the dyad, a specialised calcium microdomain formed between the t-ts and the junctional portion of the SR (jSR). We further describe how image analysis methods may be applied to interrogate the 3D volumes to provide quantitative data such as the volume of the cell occupied by the t-t and SR membranes and the volumes and surface area of jSR patches. We consider the strengths and weaknesses of the SBF-SEM technique, pitfalls in sample preparation together with tips and methods for image analysis. By providing a 'big picture' view at high resolutions, in comparison to conventional confocal microscopy, SBF-SEM represents a paradigm shift for imaging cellular networks in their native environment.
Collapse
Affiliation(s)
- Christian Pinali
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Ashraf Kitmitto
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK.
| |
Collapse
|
5
|
Vervliet T, Decrock E, Molgó J, Sorrentino V, Missiaen L, Leybaert L, De Smedt H, Kasri NN, Parys JB, Bultynck G. Bcl-2 binds to and inhibits ryanodine receptors. J Cell Sci 2014; 127:2782-92. [DOI: 10.1242/jcs.150011] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The anti-apoptotic B-cell lymphoma-2 (Bcl-2) protein not only counteracts apoptosis at the mitochondria by scaffolding pro-apoptotic Bcl-2-family members, but also acts at the endoplasmic reticulum, thereby controlling intracellular Ca2+ dynamics. Bcl-2 inhibits Ca2+ release by targeting the inositol 1,4,5-trisphosphate receptor (IP3R). Sequence analysis revealed that the Bcl-2-binding site on the IP3R displays strong homology with a conserved sequence present in all three ryanodine-receptor (RyR) isoforms. We now report that, Bcl-2 co-immunoprecipitated with RyRs in ectopic expression systems and in native rat hippocampi, indicating the existence of endogenous RyR/Bcl-2 complexes. Purified RyR domains containing the putative Bcl-2-binding site bound full-length Bcl-2 in pull-down experiments and interacted with Bcl-2's BH4 domain in surface-plasmon-resonance experiments, suggesting a direct interaction. Exogenous expression of full-length Bcl-2 or electroporation loading of Bcl-2's BH4-domain dampened RyR-mediated Ca2+ release in HEK293 cell models. Finally, introducing the BH4-domain peptide into hippocampal neurons via a patch pipette decreased RyR-mediated Ca2+ release. In conclusion, this study identifies Bcl-2 as a novel inhibitor of RyR-based intracellular Ca2+-release channels.
Collapse
|
6
|
Numata T, Ogawa N, Takahashi N, Mori Y. TRP channels as sensors of oxygen availability. Pflugers Arch 2013; 465:1075-85. [DOI: 10.1007/s00424-013-1237-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 01/31/2013] [Accepted: 01/31/2013] [Indexed: 11/28/2022]
|
7
|
Tian X, Liu Y, Liu Y, Wang R, Wagenknecht T, Liu Z, Chen SRW. Ligand-dependent conformational changes in the clamp region of the cardiac ryanodine receptor. J Biol Chem 2012; 288:4066-75. [PMID: 23258540 DOI: 10.1074/jbc.m112.427864] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Global conformational changes in the three-dimensional structure of the Ca(2+) release channel/ryanodine receptor (RyR) occur upon ligand activation. A number of ligands are able to activate the RyR channel, but whether these structurally diverse ligands induce the same or different conformational changes in the channel is largely unknown. Here we constructed a fluorescence resonance energy transfer (FRET)-based probe by inserting a CFP after residue Ser-2367 and a YFP after residue Tyr-2801 in the cardiac RyR (RyR2) to yield a CFP- and YFP-dual labeled RyR2 (RyR2(Ser-2367-CFP/Tyr-2801-YFP)). Both of these insertion sites have previously been mapped to the "clamp" region in the four corners of the square-shaped cytoplasmic assembly of the three-dimensional structure of RyR2. Using this novel FRET probe, we monitored the extent of conformational changes in the clamp region of RyR2(Ser-2367-CFP/Tyr-2801-YFP) induced by various ligands. We also monitored the extent of Ca(2+) release induced by the same ligands in HEK293 cells expressing RyR2(Ser-2367-CFP/Tyr-2801-YFP). We detected conformational changes in the clamp region for the ligands caffeine, aminophylline, theophylline, ATP, and ryanodine but not for Ca(2+) or 4-chloro-m-cresol, although they all induced Ca(2+) release. Interestingly, caffeine is able to induce further conformational changes in the clamp region of the ryanodine-modified channel, suggesting that ryanodine does not lock RyR in a fixed conformation. Our data demonstrate that conformational changes in the clamp region of RyR are ligand-dependent and suggest the existence of multiple ligand dependent RyR activation mechanisms associated with distinct conformational changes.
Collapse
Affiliation(s)
- Xixi Tian
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology and Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | | | | | | | | | | | | |
Collapse
|
8
|
Popova OB, Baker MR, Tran TP, Le T, Serysheva II. Identification of ATP-binding regions in the RyR1 Ca²⁺ release channel. PLoS One 2012; 7:e48725. [PMID: 23144945 PMCID: PMC3492408 DOI: 10.1371/journal.pone.0048725] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 09/28/2012] [Indexed: 12/11/2022] Open
Abstract
ATP is an important modulator of gating in type 1 ryanodine receptor (RyR1), also known as a Ca2+ release channel in skeletal muscle cells. The activating effect of ATP on this channel is achieved by directly binding to one or more sites on the RyR1 protein. However, the number and location of these sites have yet to be determined. To identify the ATP-binding regions within RyR1 we used 2N3ATP-2′,3′-Biotin-LC-Hydrazone (BioATP-HDZ), a photo-reactive ATP analog to covalently label the channel. We found that BioATP-HDZ binds RyR1 specifically with an IC50 = 0.6±0.2 mM, comparable with the reported EC50 for activation of RyR1 with ATP. Controlled proteolysis of labeled RyR1 followed by sequence analysis revealed three fragments with apparent molecular masses of 95, 45 and 70 kDa that were crosslinked by BioATP-HDZ and identified as RyR1 sequences. Our analysis identified four glycine-rich consensus motifs that can potentially constitute ATP-binding sites and are located within the N-terminal 95-kDa fragment. These putative nucleotide-binding sequences include amino acids 699–704, 701–706, 1081–1084 and 1195–1200, which are conserved among the three RyR isoforms. Located next to the N-terminal disease hotspot region in RyR1, these sequences may communicate the effects of ATP-binding to channel function by tuning conformational motions within the neighboring cytoplasmic regulatory domains. Two other labeled fragments lack ATP-binding consensus motifs and may form non-canonical ATP-binding sites. Based on domain topology in the 3D structure of RyR1 it is also conceivable that the identified ATP-binding regions, despite their wide separation in the primary sequence, may actually constitute the same non-contiguous ATP-binding pocket within the channel tetramer.
Collapse
Affiliation(s)
- Olga B. Popova
- Department of Biochemistry and Molecular Biology, The University of Texas at Houston Medical School, Houston, Texas, United States of America
| | - Mariah R. Baker
- Department of Biochemistry and Molecular Biology, The University of Texas at Houston Medical School, Houston, Texas, United States of America
| | - Tina P. Tran
- Department of Biochemistry and Molecular Biology, The University of Texas at Houston Medical School, Houston, Texas, United States of America
| | - Tri Le
- Department of Biochemistry and Molecular Biology, The University of Texas at Houston Medical School, Houston, Texas, United States of America
| | - Irina I. Serysheva
- Department of Biochemistry and Molecular Biology, The University of Texas at Houston Medical School, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
9
|
Raina SA, Tsai J, Samsó M, Fessenden JD. FRET-based localization of fluorescent protein insertions within the ryanodine receptor type 1. PLoS One 2012; 7:e38594. [PMID: 22719904 PMCID: PMC3374828 DOI: 10.1371/journal.pone.0038594] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/11/2012] [Indexed: 11/21/2022] Open
Abstract
Fluorescent protein (FP) insertions have often been used to localize primary structure elements in mid-resolution 3D cryo electron microscopic (EM) maps of large protein complexes. However, little is known as to the precise spatial relationship between the location of the fused FP and its insertion site within a larger protein. To gain insights into these structural considerations, Förster resonance energy transfer (FRET) measurements were used to localize green fluorescent protein (GFP) insertions within the ryanodine receptor type 1 (RyR1), a large intracellular Ca(2+) release channel that plays a key role in skeletal muscle excitation contraction coupling. A series of full-length His-tagged GFP-RyR1 fusion constructs were created, expressed in human embryonic kidney (HEK)-293T cells and then complexed with Cy3NTA, a His-tag specific FRET acceptor. FRET efficiency values measured from each GFP donor to Cy3NTA bound to each His tag acceptor site were converted into intermolecular distances and the positions of each inserted GFP were then triangulated relative to a previously published X-ray crystal structure of a 559 amino acid RyR1 fragment. We observed that the chromophoric centers of fluorescent proteins inserted into RyR1 can be located as far as 45 Å from their insertion sites and that the fused proteins can also be located in internal cavities within RyR1. These findings should prove useful in interpreting structural results obtained in cryo EM maps using fusions of small fluorescent proteins. More accurate point-to-point distance information may be obtained using complementary orthogonal labeling systems that rely on fluorescent probes that bind directly to amino acid side chains.
Collapse
Affiliation(s)
- Shweta A. Raina
- Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
| | - Jeffrey Tsai
- Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
| | - Montserrat Samsó
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - James D. Fessenden
- Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
| |
Collapse
|
10
|
Techniques and Methodologies to Study the Ryanodine Receptor at the Molecular, Subcellular and Cellular Level. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:183-215. [DOI: 10.1007/978-94-007-2888-2_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
11
|
The structural biology of ryanodine receptors. SCIENCE CHINA-LIFE SCIENCES 2011; 54:712-24. [DOI: 10.1007/s11427-011-4198-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 05/30/2011] [Indexed: 10/18/2022]
|
12
|
Suetomi T, Yano M, Uchinoumi H, Fukuda M, Hino A, Ono M, Xu X, Tateishi H, Okuda S, Doi M, Kobayashi S, Ikeda Y, Yamamoto T, Ikemoto N, Matsuzaki M. Mutation-linked defective interdomain interactions within ryanodine receptor cause aberrant Ca²⁺release leading to catecholaminergic polymorphic ventricular tachycardia. Circulation 2011; 124:682-94. [PMID: 21768539 DOI: 10.1161/circulationaha.111.023259] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The molecular mechanism by which catecholaminergic polymorphic ventricular tachycardia is induced by single amino acid mutations within the cardiac ryanodine receptor (RyR2) remains elusive. In the present study, we investigated mutation-induced conformational defects of RyR2 using a knockin mouse model expressing the human catecholaminergic polymorphic ventricular tachycardia-associated RyR2 mutant (S2246L; serine to leucine mutation at the residue 2246). METHODS AND RESULTS All knockin mice we examined produced ventricular tachycardia after exercise on a treadmill. cAMP-dependent increase in the frequency of Ca²⁺ sparks was more pronounced in saponin-permeabilized knockin cardiomyocytes than in wild-type cardiomyocytes. Site-directed fluorescent labeling and quartz microbalance assays of the specific binding of DP2246 (a peptide corresponding to the 2232 to 2266 region: the 2246 domain) showed that DP2246 binds with the K201-binding sequence of RyR2 (1741 to 2270). Introduction of S2246L mutation into the DP2246 increased the affinity of peptide binding. Fluorescence quench assays of interdomain interactions within RyR2 showed that tight interaction of the 2246 domain/K201-binding domain is coupled with domain unzipping of the N-terminal (1 to 600)/central (2000 to 2500) domain pair in an allosteric manner. Dantrolene corrected the mutation-caused domain unzipping of the domain switch and stopped the exercise-induced ventricular tachycardia. CONCLUSIONS The catecholaminergic polymorphic ventricular tachycardia-linked mutation of RyR2, S2246L, causes an abnormally tight local subdomain-subdomain interaction within the central domain involving the mutation site, which induces defective interaction between the N-terminal and central domains. This results in an erroneous activation of Ca²⁺ channel in a diastolic state reflecting on the increased Ca²⁺ spark frequency, which then leads to lethal arrhythmia.
Collapse
Affiliation(s)
- Takeshi Suetomi
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Song DW, Lee JG, Youn HS, Eom SH, Kim DH. Ryanodine receptor assembly: A novel systems biology approach to 3D mapping. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2011; 105:145-61. [DOI: 10.1016/j.pbiomolbio.2010.09.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 09/14/2010] [Accepted: 09/28/2010] [Indexed: 10/19/2022]
|
14
|
Wang R, Zhong X, Meng X, Koop A, Tian X, Jones PP, Fruen BR, Wagenknecht T, Liu Z, Chen SRW. Localization of the dantrolene-binding sequence near the FK506-binding protein-binding site in the three-dimensional structure of the ryanodine receptor. J Biol Chem 2011; 286:12202-12. [PMID: 21262961 DOI: 10.1074/jbc.m110.194316] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dantrolene is believed to stabilize interdomain interactions between the NH2-terminal and central regions of ryanodine receptors by binding to the NH2-terminal residues 590-609 in skeletal ryanodine receptor (RyR1) and residues 601-620 in cardiac ryanodine receptor (RyR2). To gain further insight into the structural basis of dantrolene action, we have attempted to localize the dantrolene-binding sequence in RyR1/RyR2 by using GFP as a structural marker and three-dimensional cryo-EM. We inserted GFP into RyR2 after residues Arg-626 and Tyr-846 to generate GFP-RyR2 fusion proteins, RyR2Arg-626-GFP and RyR2Tyr-846-GFP. Insertion of GFP after residue Arg-626 abolished the binding of a bulky GST- or cyan fluorescent protein-tagged FKBP12.6 but not the binding of a smaller, nontagged FKBP12.6, suggesting that residue Arg-626 and the dantrolene-binding sequence are located near the FKBP12.6-binding site. Using cryo-EM, we have mapped the three-dimensional location of Tyr-846-GFP to domain 9, which is also adjacent to the FKBP12.6-binding site. To further map the three-dimensional location of the dantrolene-binding sequence, we generated 10 FRET pairs based on four known three-dimensional locations (FKBP12.6, Ser-437-GFP, Tyr-846-GFP, and Ser-2367-GFP). Based on the FRET efficiencies of these FRET pairs and the corresponding distance relationships, we mapped the three-dimensional location of Arg-626-GFP or -cyan fluorescent protein, hence the dantrolene-binding sequence, to domain 9 near the FKBP12.6-binding site but distant to the central region around residue Ser-2367. An allosteric mechanism by which dantrolene stabilizes interdomain interactions between the NH2-terminal and central regions is proposed.
Collapse
Affiliation(s)
- Ruiwu Wang
- Libin Cardiovascular Institute of Alberta, Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Lanner JT, Georgiou DK, Joshi AD, Hamilton SL. Ryanodine receptors: structure, expression, molecular details, and function in calcium release. Cold Spring Harb Perspect Biol 2010; 2:a003996. [PMID: 20961976 DOI: 10.1101/cshperspect.a003996] [Citation(s) in RCA: 546] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ryanodine receptors (RyRs) are located in the sarcoplasmic/endoplasmic reticulum membrane and are responsible for the release of Ca(2+) from intracellular stores during excitation-contraction coupling in both cardiac and skeletal muscle. RyRs are the largest known ion channels (> 2MDa) and exist as three mammalian isoforms (RyR 1-3), all of which are homotetrameric proteins that interact with and are regulated by phosphorylation, redox modifications, and a variety of small proteins and ions. Most RyR channel modulators interact with the large cytoplasmic domain whereas the carboxy-terminal portion of the protein forms the ion-conducting pore. Mutations in RyR2 are associated with human disorders such as catecholaminergic polymorphic ventricular tachycardia whereas mutations in RyR1 underlie diseases such as central core disease and malignant hyperthermia. This chapter examines the current concepts of the structure, function and regulation of RyRs and assesses the current state of understanding of their roles in associated disorders.
Collapse
Affiliation(s)
- Johanna T Lanner
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, Houston, Texas 77030,USA
| | | | | | | |
Collapse
|
16
|
Wagenknecht TC, Liu Z. Electron microscopy of ryanodine receptors. CURRENT TOPICS IN MEMBRANES 2010; 66:27-47. [PMID: 22353475 DOI: 10.1016/s1063-5823(10)66002-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Terence C Wagenknecht
- Wadsworth Center, New York State Department of Health, Albany, New York, USA; Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York, USA
| | | |
Collapse
|
17
|
Liu Z, Wang R, Tian X, Zhong X, Gangopadhyay J, Cole R, Ikemoto N, Chen SRW, Wagenknecht T. Dynamic, inter-subunit interactions between the N-terminal and central mutation regions of cardiac ryanodine receptor. J Cell Sci 2010; 123:1775-84. [PMID: 20427316 PMCID: PMC2864716 DOI: 10.1242/jcs.064071] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2010] [Indexed: 11/20/2022] Open
Abstract
Naturally occurring mutations in the cardiac ryanodine receptor (RyR2) have been linked to certain types of cardiac arrhythmias and sudden death. Two mutation hotspots that lie in the N-terminal and central regions of RyR2 are predicted to interact with one another and to form an important channel regulator switch. To monitor the conformational dynamics involving these regions, we generated a fluorescence resonance energy transfer (FRET) pair. A yellow fluorescent protein (YFP) was inserted into RyR2 after residue Ser437 in the N-terminal region, and a cyan fluorescent protein (CFP) was inserted after residue Ser2367 in the central region, to form a dual YFP- and CFP-labeled RyR2 (RyR2(S437-YFP/S2367-CFP)). We transfected HEK293 cells with RyR2(S437-YFP/S2367-CFP) cDNAs, and then examined them by using confocal microscopy and by measuring the FRET signal in live cells. The FRET signals are influenced by modulators of RyR2, by domain peptides that mimic the effects of disease causing RyR2 mutations, and by various drugs. Importantly, FRET signals were also readily detected in cells co-transfected with single CFP (RyR2(S437-YFP)) and single YFP (RyR2(S2367-CFP)) labeled RyR2, indicating that the interaction between the N-terminal and central mutation regions is an inter-subunit interaction. Our studies demonstrate that FRET analyses of this CFP- and YFP-labeled RyR2 can be used not only for investigating the conformational dynamics associated with RyR2 channel gating, but potentially, also for identifying drugs that are capable of stabilizing the conformations of RyR2.
Collapse
Affiliation(s)
- Zheng Liu
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ono M, Yano M, Hino A, Suetomi T, Xu X, Susa T, Uchinoumi H, Tateishi H, Oda T, Okuda S, Doi M, Kobayashi S, Yamamoto T, Koseki N, Kyushiki H, Ikemoto N, Matsuzaki M. Dissociation of calmodulin from cardiac ryanodine receptor causes aberrant Ca(2+) release in heart failure. Cardiovasc Res 2010; 87:609-17. [PMID: 20388639 DOI: 10.1093/cvr/cvq108] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Calmodulin (CaM) is well known to modulate the channel function of the cardiac ryanodine receptor (RyR2). However, the possible role of CaM on the aberrant Ca(2+) release in diseased hearts remains unclear. In this study, we investigated the state of RyR2-bound CaM and channel dysfunctions in pacing-induced failing hearts. METHODS AND RESULTS The characteristics of CaM binding to RyR2 and the role of CaM on the aberrant Ca(2+) release were assessed in normal and failing canine hearts. The affinity of CaM binding to RyR2 was lower in failing sarcoplasmic reticulum (SR) than in normal SR. Addition of FK506, which dissociates FKBP12.6 from RyR2, to normal SR reduced the CaM-binding affinity. Dantrolene restored a normal level of the CaM-binding affinity in either FK506-treated (normal) SR or failing SR, suggesting that the defective inter-domain interaction between the N-terminal domain and the central domain of RyR2 (the therapeutic target of dantrolene) is involved in the reduction of the CaM-binding affinity in failing hearts. In saponin-permeabilized cardiomyocytes, the frequency of spontaneous Ca(2+) sparks was much more increased in failing cardiomyocytes than in normal cardiomyocytes, whereas the addition of a high concentration of CaM attenuated the aberrant increase of Ca(2+) sparks. CONCLUSION The defective inter-domain interaction between N-terminal and central domains within RyR2 reduces the binding affinity of CaM to RyR2, thereby causing the spontaneous Ca(2+) release events in failing hearts. Correction of the defective CaM binding may be a new strategy to protect against the aberrant Ca(2+) release in heart failure.
Collapse
Affiliation(s)
- Makoto Ono
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Uchinoumi H, Yano M, Suetomi T, Ono M, Xu X, Tateishi H, Oda T, Okuda S, Doi M, Kobayashi S, Yamamoto T, Ikeda Y, Ohkusa T, Ikemoto N, Matsuzaki M. Catecholaminergic polymorphic ventricular tachycardia is caused by mutation-linked defective conformational regulation of the ryanodine receptor. Circ Res 2010; 106:1413-24. [PMID: 20224043 DOI: 10.1161/circresaha.109.209312] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by a single point mutation in a well-defined region of the cardiac type 2 ryanodine receptor (RyR)2. However, the underlying mechanism by which a single mutation in such a large molecule produces drastic effects on channel function remains unresolved. OBJECTIVE Using a knock-in (KI) mouse model with a human CPVT-associated RyR2 mutation (R2474S), we investigated the molecular mechanism by which CPVT is induced by a single point mutation within the RyR2. METHODS AND RESULTS The R2474S/+ KI mice showed no apparent structural or histological abnormalities in the heart, but they showed clear indications of other abnormalities. Bidirectional or polymorphic ventricular tachycardia was induced after exercise on a treadmill. The interaction between the N-terminal (amino acids 1 to 600) and central (amino acids 2000 to 2500) domains of the RyR2 (an intrinsic mechanism to close Ca(2+) channels) was weakened (domain unzipping). On protein kinase A-mediated phosphorylation of the RyR2, this domain unzipping further increased, resulting in a significant increase in the frequency of spontaneous Ca(2+) transients. cAMP-induced aberrant Ca(2+) release events (Ca(2+) sparks/waves) occurred at much lower sarcoplasmic reticulum Ca(2+) content as compared to the wild type. Addition of a domain-unzipping peptide, DPc10 (amino acids 2460 to 2495), to the wild type reproduced the aforementioned abnormalities that are characteristic of the R2474S/+ KI mice. Addition of DPc10 to the (cAMP-treated) KI cardiomyocytes produced no further effect. CONCLUSIONS A single point mutation within the RyR2 sensitizes the channel to agonists and reduces the threshold of luminal [Ca(2+)] for activation, primarily mediated by defective interdomain interaction within the RyR2.
Collapse
Affiliation(s)
- Hitoshi Uchinoumi
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Xu X, Yano M, Uchinoumi H, Hino A, Suetomi T, Ono M, Tateishi H, Oda T, Okuda S, Doi M, Kobayashi S, Yamamoto T, Ikeda Y, Ikemoto N, Matsuzaki M. Defective calmodulin binding to the cardiac ryanodine receptor plays a key role in CPVT-associated channel dysfunction. Biochem Biophys Res Commun 2010; 394:660-6. [PMID: 20226167 DOI: 10.1016/j.bbrc.2010.03.046] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Accepted: 03/07/2010] [Indexed: 10/19/2022]
Abstract
Calmodulin (CaM), one of the accessory proteins of the cardiac ryanodine receptor (RyR2), is known to play a significant role in the channel regulation of the RyR2. However, the possible involvement of calmodulin in the pathogenic process of catecholaminergic polymorphic ventricular tachycardia (CPVT) has not been investigated. In this study, we investigated the state of RyR2-bound CaM and channel dysfunctions using a knock-in (KI) mouse model with CPVT-linked RyR2 mutation (R2474S). Without added effectors, the affinity of CaM binding to the RyR2 was indistinguishable between KI and WT hearts. In response to cAMP (1 micromol/L), the RyR2 phosphorylation at Ser2808 increased in both WT and KI hearts to the same extent. However, cAMP caused a significant decrease of the CaM-binding affinity in KI hearts, but the affinity was unchanged in WT. Dantrolene restored a normal level of CaM-binding affinity in the cAMP-treated KI hearts, suggesting that defective inter-domain interaction between the N-terminal domain and the central domain of the RyR2 (the target of therapeutic effect of dantrolene) is involved in the cAMP-induced reduction of the CaM-binding affinity. In saponin-permeabilized cardiomyocytes, the addition of cAMP increased the frequency of spontaneous Ca(2+) sparks to a significantly larger extent in KI cardiomyocytes than in WT cardiomyocytes, whereas the addition of a high concentration of CaM attenuated the aberrant increase of Ca(2+) sparks. In conclusion, CPVT mutation causes defective inter-domain interaction, significant reduction in the ability of CaM binding to the RyR2, spontaneous Ca(2+) leak, and then lethal arrhythmia.
Collapse
Affiliation(s)
- Xiaojuan Xu
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Thomas NL, Maxwell C, Mukherjee S, Williams AJ. Ryanodine receptor mutations in arrhythmia: The continuing mystery of channel dysfunction. FEBS Lett 2010; 584:2153-60. [PMID: 20132818 DOI: 10.1016/j.febslet.2010.01.057] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 01/27/2010] [Accepted: 01/29/2010] [Indexed: 11/13/2022]
Abstract
Mutations in RyR2 are causative of an inherited disorder which often results in sudden cardiac death. Dysfunctional channel behaviour has been the subject of many investigations varying from single channel analysis through to complex animal models. This review discusses recent advances in the field, describes the controversy surrounding the exact consequences of RyR2 mutation and how the disparate data may be reconciled. This heterogeneity of function with respect to the effects of polymorphisms, phosphorylation, cytosolic and luminal Ca(2+) as well as inter-domain interactions may have important implications for the recent pharmaceutical therapies which have been put forward. We surmise that a comprehensive characterisation of mutations on a case-by-case basis may be beneficial for the development of specifically targeted therapies.
Collapse
Affiliation(s)
- N Lowri Thomas
- Department of Cardiology, Wales Heart Research Institute, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| | | | | | | |
Collapse
|
22
|
Medeiros-Domingo A, Bhuiyan ZA, Tester DJ, Hofman N, Bikker H, van Tintelen JP, Mannens MM, Wilde AA, Ackerman MJ. The RYR2-encoded ryanodine receptor/calcium release channel in patients diagnosed previously with either catecholaminergic polymorphic ventricular tachycardia or genotype negative, exercise-induced long QT syndrome: a comprehensive open reading frame mutational analysis. J Am Coll Cardiol 2009; 54:2065-74. [PMID: 19926015 PMCID: PMC2880864 DOI: 10.1016/j.jacc.2009.08.022] [Citation(s) in RCA: 257] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 08/28/2009] [Accepted: 08/30/2009] [Indexed: 12/20/2022]
Abstract
OBJECTIVES This study was undertaken to determine the spectrum and prevalence of mutations in the RYR2-encoded cardiac ryanodine receptor in cases with exertional syncope and normal corrected QT interval (QTc). BACKGROUND Mutations in RYR2 cause type 1 catecholaminergic polymorphic ventricular tachycardia (CPVT1), a cardiac channelopathy with increased propensity for lethal ventricular dysrhythmias. Most RYR2 mutational analyses target 3 canonical domains encoded by <40% of the translated exons. The extent of CPVT1-associated mutations localizing outside of these domains remains unknown as RYR2 has not been examined comprehensively in most patient cohorts. METHODS Mutational analysis of all RYR2 exons was performed using polymerase chain reaction, high-performance liquid chromatography, and deoxyribonucleic acid sequencing on 155 unrelated patients (49% females, 96% Caucasian, age at diagnosis 20 +/- 15 years, mean QTc 428 +/- 29 ms), with either clinical diagnosis of CPVT (n = 110) or an initial diagnosis of exercise-induced long QT syndrome but with QTc <480 ms and a subsequent negative long QT syndrome genetic test (n = 45). RESULTS Sixty-three (34 novel) possible CPVT1-associated mutations, absent in 400 reference alleles, were detected in 73 unrelated patients (47%). Thirteen new mutation-containing exons were identified. Two-thirds of the CPVT1-positive patients had mutations that localized to 1 of 16 exons. CONCLUSIONS Possible CPVT1 mutations in RYR2 were identified in nearly one-half of this cohort; 45 of the 105 translated exons are now known to host possible mutations. Considering that approximately 65% of CPVT1-positive cases would be discovered by selective analysis of 16 exons, a tiered targeting strategy for CPVT genetic testing should be considered.
Collapse
Affiliation(s)
| | - Zahurul A. Bhuiyan
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Netherlands
| | - David J. Tester
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester MN USA
| | - Nynke Hofman
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Netherlands
| | - Hennie Bikker
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Netherlands
| | - J Peter van Tintelen
- Department of Genetics, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Marcel M.A.M Mannens
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Netherlands
| | - Arthur A.M. Wilde
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Netherlands
- Department of Cardiology and Heart Failure Research Centre, Academic Medical Center, University of Amsterdam, Netherlands
| | - Michael J. Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester MN USA
- Department of Medicine/Division of Cardiovascular Diseases, Mayo Clinic, Rochester MN USA
- Department of Pediatrics/Division of Pediatric Cardiology, Mayo Clinic, Rochester MN USA
| |
Collapse
|
23
|
Fessenden JD. Förster resonance energy transfer measurements of ryanodine receptor type 1 structure using a novel site-specific labeling method. PLoS One 2009; 4:e7338. [PMID: 19823671 PMCID: PMC2757896 DOI: 10.1371/journal.pone.0007338] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 09/14/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND While the static structure of the intracellular Ca(2+) release channel, the ryanodine receptor type 1 (RyR1) has been determined using cryo electron microscopy, relatively little is known concerning changes in RyR1 structure that accompany channel gating. Förster resonance energy transfer (FRET) methods can resolve small changes in protein structure although FRET measurements of RyR1 are hampered by an inability to site-specifically label the protein with fluorescent probes. METHODOLOGY/PRINCIPAL FINDINGS A novel site-specific labeling method is presented that targets a FRET acceptor, Cy3NTA to 10-residue histidine (His) tags engineered into RyR1. Cy3NTA, comprised of the fluorescent dye Cy3, coupled to two Ni(2+)/nitrilotriacetic acid moieties, was synthesized and functionally tested for binding to His-tagged green fluorescent protein (GFP). GFP fluorescence emission and Cy3NTA absorbance spectra overlapped significantly, indicating that FRET could occur (Förster distance = 6.3 nm). Cy3NTA bound to His(10)-tagged GFP, quenching its fluorescence by 88%. GFP was then fused to the N-terminus of RyR1 and His(10) tags were placed either at the N-terminus of the fused GFP or between GFP and RyR1. Cy3NTA reduced fluorescence of these fusion proteins by 75% and this quenching could be reversed by photobleaching Cy3, thus confirming GFP-RyR1 quenching via FRET. A His(10) tag was then placed at amino acid position 1861 and FRET was measured from GFP located at either the N-terminus or at position 618 to Cy3NTA bound to this His tag. While minimal FRET was detected between GFP at position 1 and Cy3NTA at position 1861, 53% energy transfer was detected from GFP at position 618 to Cy3NTA at position 1861, thus indicating that these sites are in close proximity to each other. CONCLUSIONS/SIGNIFICANCE These findings illustrate the potential of this site-specific labeling system for use in future FRET-based experiments to elucidate novel aspects of RyR1 structure.
Collapse
Affiliation(s)
- James D Fessenden
- Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America.
| |
Collapse
|
24
|
Blayney LM, Lai FA. Ryanodine receptor-mediated arrhythmias and sudden cardiac death. Pharmacol Ther 2009; 123:151-77. [PMID: 19345240 PMCID: PMC2704947 DOI: 10.1016/j.pharmthera.2009.03.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 03/03/2009] [Indexed: 12/25/2022]
Abstract
The cardiac ryanodine receptor-Ca2+ release channel (RyR2) is an essential sarcoplasmic reticulum (SR) transmembrane protein that plays a central role in excitation–contraction coupling (ECC) in cardiomyocytes. Aberrant spontaneous, diastolic Ca2+ leak from the SR due to dysfunctional RyR2 contributes to the formation of delayed after-depolarisations, which are thought to underlie the fatal arrhythmia that occurs in both heart failure (HF) and in catecholaminergic polymorphic ventricular tachycardia (CPVT). CPVT is an inherited disorder associated with mutations in either the RyR2 or a SR luminal protein, calsequestrin. RyR2 shows normal function at rest in CPVT but the RyR2 dysfunction is unmasked by physical exercise or emotional stress, suggesting abnormal RyR2 activation as an underlying mechanism. Several potential mechanisms have been advanced to explain the dysfunctional RyR2 observed in HF and CPVT, including enhanced RyR2 phosphorylation status, altered RyR2 regulation at luminal/cytoplasmic sites and perturbed RyR2 intra/inter-molecular interactions. This review considers RyR2 dysfunction in the context of the structural and functional modulation of the channel, and potential therapeutic strategies to stabilise RyR2 function in cardiac pathology.
Collapse
Affiliation(s)
- Lynda M Blayney
- Wales Heart Research Institute, Cardiff University School of Medicine, Cardiff CF144XN, UK.
| | | |
Collapse
|
25
|
Laver DR. Luminal Ca(2+) activation of cardiac ryanodine receptors by luminal and cytoplasmic domains. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2009; 39:19-26. [PMID: 19255753 DOI: 10.1007/s00249-009-0417-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 01/28/2009] [Accepted: 02/02/2009] [Indexed: 10/21/2022]
Abstract
The ryanodine receptors form the calcium release channel in the membrane of the sarcoplasmic reticulum (SR, the main intracellular Ca(2+) store). The importance of ryanodine receptors (RyRs) to cardiac pacemaking and rhythmicity is highlighted by more than 69 mutations, RyR mutations, which underlie arrhythmias and sudden cardiac death. Although most of these mutations lie in cytoplasmic domains, they all cause increased RyR activation by Ca(2+) in the SR lumen. Presented here is a review of the mechanisms by which cytoplasmic domains of the RyR can determine luminal activation.
Collapse
Affiliation(s)
- Derek R Laver
- School of Biomedical Sciences, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
26
|
Affiliation(s)
- Susan L Hamilton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
27
|
Meng X, Wang G, Viero C, Wang Q, Mi W, Su XD, Wagenknecht T, Williams AJ, Liu Z, Yin CC. CLIC2-RyR1 interaction and structural characterization by cryo-electron microscopy. J Mol Biol 2009; 387:320-34. [PMID: 19356589 DOI: 10.1016/j.jmb.2009.01.059] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2008] [Revised: 01/18/2009] [Accepted: 01/27/2009] [Indexed: 11/18/2022]
Abstract
Chloride intracellular channel 2 (CLIC2), a newly discovered small protein distantly related to the glutathione transferase (GST) structural family, is highly expressed in cardiac and skeletal muscle, although its physiological function in these tissues has not been established. In the present study, [3H]ryanodine binding, Ca2+ efflux from skeletal sarcoplasmic reticulum (SR) vesicles, single channel recording, and cryo-electron microscopy were employed to investigate whether CLIC2 can interact with skeletal ryanodine receptor (RyR1) and modulate its channel activity. We found that: (1) CLIC2 facilitated [3H]ryanodine binding to skeletal SR and purified RyR1, by increasing the binding affinity of ryanodine for its receptor without significantly changing the apparent maximal binding capacity; (2) CLIC2 reduced the maximal Ca2+ efflux rate from skeletal SR vesicles; (3) CLIC2 decreased the open probability of RyR1 channel, through increasing the mean closed time of the channel; (4) CLIC2 bound to a region between domains 5 and 6 in the clamp-shaped region of RyR1; (5) and in the same clamp region, domains 9 and 10 became separated after CLIC2 binding, indicating CLIC2 induced a conformational change of RyR1. These data suggest that CLIC2 can interact with RyR1 and modulate its channel activity. We propose that CLIC2 functions as an intrinsic stabilizer of the closed state of RyR channels.
Collapse
Affiliation(s)
- Xing Meng
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yano M, Yamamoto T, Kobayashi S, Matsuzaki M. Role of ryanodine receptor as a Ca²(+) regulatory center in normal and failing hearts. J Cardiol 2008; 53:1-7. [PMID: 19167631 DOI: 10.1016/j.jjcc.2008.10.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2008] [Accepted: 10/20/2008] [Indexed: 11/19/2022]
Abstract
Abnormal Ca²(+) cycling is important in various cardiac diseases. Evidence has accumulated that dysregulation of Ca²(+) release from the ryanodine receptor (RyR2) plays a significant role in cardiac dysfunction. Spontaneous Ca²(+) release through RyR2 during diastole decreases sarcoplasmic reticulum (SR) Ca²(+) content, and also induces delayed after depolarization (DAD) as a substrate for lethal arrhythmia. Several disease-linked mutations in the RyR have been reported in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT) or arrythmogenic right ventricular cardiomyopathy type 2 (ARVC2). The unique distribution of these mutation sites has produced the concept that the interaction among the putative regulatory domains within the RyR may play a key role in regulating the channel opening, and that there seems to be a common abnormality in the channel disorder between heart failure and CPVT/ARVC2. We review here the considerable body of evidence regarding defective channel gating of RyR2 in the pathogenesis of heart failure and lethal arrhythmia.
Collapse
Affiliation(s)
- Masafumi Yano
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | | | | | | |
Collapse
|
29
|
Yu Z, Holst MJ, Hayashi T, Bajaj CL, Ellisman MH, McCammon JA, Hoshijima M. Three-dimensional geometric modeling of membrane-bound organelles in ventricular myocytes: bridging the gap between microscopic imaging and mathematical simulation. J Struct Biol 2008; 164:304-13. [PMID: 18835449 DOI: 10.1016/j.jsb.2008.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 09/09/2008] [Accepted: 09/10/2008] [Indexed: 10/21/2022]
Abstract
A general framework of image-based geometric processing is presented to bridge the gap between three-dimensional (3D) imaging that provides structural details of a biological system and mathematical simulation where high-quality surface or volumetric meshes are required. A 3D density map is processed in the order of image pre-processing (contrast enhancement and anisotropic filtering), feature extraction (boundary segmentation and skeletonization), and high-quality and realistic surface (triangular) and volumetric (tetrahedral) mesh generation. While the tool-chain described is applicable to general types of 3D imaging data, the performance is demonstrated specifically on membrane-bound organelles in ventricular myocytes that are imaged and reconstructed with electron microscopic (EM) tomography and two-photon microscopy (T-PM). Of particular interest in this study are two types of membrane-bound Ca(2+)-handling organelles, namely, transverse tubules (T-tubules) and junctional sarcoplasmic reticulum (jSR), both of which play an important role in regulating the excitation-contraction (E-C) coupling through dynamic Ca(2+) mobilization in cardiomyocytes.
Collapse
Affiliation(s)
- Zeyun Yu
- Department of Mathematics, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Yano M, Yamamoto T, Kobayashi S, Ikeda Y, Matsuzaki M. Defective Ca2+ cycling as a key pathogenic mechanism of heart failure. Circ J 2008; 72 Suppl A:A22-30. [PMID: 18772523 DOI: 10.1253/circj.cj-08-0070] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Structural and functional alterations in the Ca(2+) regulatory proteins present in the sarcoplasmic reticulum (SR) have recently been shown to play a crucial role in the pathogenesis of heart failure (HF), and lethal arrhythmia as well. Chronic activation of the sympathetic nervous system induces abnormalities in both the function and structure of these proteins. For instance, the diastolic Ca(2+) leak through the SR Ca(2+) release channel (ryanodine receptor) reduces the SR Ca(2+) content, inducing contractile dysfunction. Moreover, the Ca(2+) leak provides a substrate for delayed after depolarization that leads to lethal arrhythmia. There is a considerable body of evidence regarding the role of Ca(2+) cycling abnormality in HF.
Collapse
Affiliation(s)
- Masafumi Yano
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, Ube, Japan.
| | | | | | | | | |
Collapse
|
31
|
Abstract
Abnormal intracellular Ca(2+) handling by the sarcoplasmic reticulum (SR) is a critical factor in the development of heart failure (HF). Not only decreased Ca(2+) uptake, but also uncoordinated Ca(2+) release plays a significant role in contractile and relaxation dysfunction. Spontaneous Ca(2+) release through ryanodine receptor (RyR) 2, a huge tetrameric protein, during diastole leads to a decrease in the SR Ca(2+) content, and also triggers delayed after depolarization that is a substrate for lethal arrhythmia. Several disease-linked mutations of RyR have been reported in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT) or arrhythmogenic right ventricular cardiomyopathy type 2 (ARVC2). The unique distribution of these mutation sites has lead to the concept that an interaction among the putative regulatory domains within RyR may play a key role in regulating channel opening, and that there seems to be a common abnormality in the channel disorder of HF and CPVT/ARVC2. Recent knowledge gained from pathological conditions may lead to the development of a new therapeutic strategy for the treatment of HF or cardiac arrhythmia.
Collapse
Affiliation(s)
- Masafumi Yano
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube 755-8505, Japan.
| |
Collapse
|
32
|
Structural and functional characterization of ryanodine receptor-natrin toxin interaction. Biophys J 2008; 95:4289-99. [PMID: 18658224 DOI: 10.1529/biophysj.108.137224] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cysteine-rich secretory proteins (CRISPs) are widely distributed, and notably occur in the mammalian reproductive tract and in the salivary glands of venomous reptiles. Most CRISPs can inhibit ion channels, such as the cyclic nucleotide-gated ion channel, potassium channel, and calcium channel. Natrin is a CRISP that has been purified from snake venom. Its targets include the calcium-activated potassium channel, the voltage-gated potassium channel, and the calcium release channel/ryanodine receptor (RyR). Immunoprecipitation experiments showed that natrin binds specifically to type 1 RyR (RyR1) from skeletal muscle. Natrin was found to inhibit both the binding of ryanodine to RyR1, and the calcium-channel activity of RyR1. Cryo-electron microscopy and single-particle image reconstruction analysis revealed that natrin binds to the clamp domains of RyR1. Docking of the crystal structure of natrin into our cryo-electron microscopy density map of the RyR1 + natrin complex suggests that natrin inhibits RyR1 by stabilizing a domain-domain interaction, and that the cysteine-rich domain of natrin is crucial for binding. These findings help reveal how natrin toxin inhibits the RyR calcium release channel, and they allow us to posit a generalized mechanism that governs the interaction between CRISPs and ion channels.
Collapse
|
33
|
Localization of PKA phosphorylation site, Ser(2030), in the three-dimensional structure of cardiac ryanodine receptor. Biochem J 2008; 410:261-70. [PMID: 17967164 DOI: 10.1042/bj20071257] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PKA (protein kinase A)-dependent phosphorylation of the cardiac Ca2+-release channel/RyR2 (type 2 ryanodine receptor)is believed to directly dissociate FKBP12.6 (12.6 kDa FK506-binding protein) from the channel, causing abnormal channel activation and Ca2+ release. To gain insight into the structural basis of the regulation of RyR2 by PKA, we determined the three-dimensional location of the PKA site Ser2030. GFP (green fluorescent protein) was inserted into RyR2-wt (wild-type RyR2)and RyR2 mutant, A4860G, after Thr2023. The resultant GFP-RyR2 fusion proteins, RyR2T2023-GFP and RyR2(A4860G)T2023-GFP, were expressed in HEK-293 (human embryonic kidney) cells and functionally characterized. Ca2+-release assays revealed that both GFP-RyR2 fusion proteins formed caffeine- and ryanodine-sensitive Ca2+-release channels. Further analyses using[3H]ryanodine binding demonstrated that the insertion of GFPinto RyR2-wt after Thr2023 reduced the sensitivity of the channelto activation by Ca2+ or caffeine. RyR2(A4860G)T2023-GFP was found to be structurally more stable than RyR2T2023-GFP and was subsequently used as a basis for three-dimensional reconstruction. Cryo-electronmicroscopy and single particle image processing of the purified RyR2(A4860G)T2023-GFP protein revealed the location of the inserted GFP, and hence the Ser2030 PKA site in domain 4,a region that may be involved in signal transduction between the transmembrane and cytoplasmic domains. Like the Ser2808 PKA site reported previously, the Ser2030 site is not located close to the FKBP12.6-binding site mapped previously, indicating that neither of these PKA sites is directly involved in FKBP12.6 binding. On the basis of the three-dimensional localizations of a number of residues or regions, a model for the subunit organization in the structure of RyR2 is proposed.
Collapse
|
34
|
ARVC-related mutations in divergent region 3 alter functional properties of the cardiac ryanodine receptor. Biophys J 2008; 94:4668-77. [PMID: 18326664 PMCID: PMC2397357 DOI: 10.1529/biophysj.107.122382] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Two single-nucleotide polymorphisms in the type 2 ryanodine receptor (RyR2) leading to the nonsynonymous amino acid replacements G1885E and G1886S are associated with arrhythmogenic right ventricular cardiomyopathy in patients who are carrying both of the corresponding RyR2 alleles. The functional properties of HEK293 cell lines isogenically expressing RyR2 mutants associated with arrhythmogenic right ventricular cardiomyopathy, RyR2-G1885E, RyR2-G1886S, RyR2-G1886D (mimicking a constitutively phosphorylated Ser(1886)), and the double mutant RyR2-G1885E/G1886S were investigated by analyzing the intracellular Ca(2+) release activity resulting from store-overload-induced calcium release. The substitution of serine for Gly(1886) caused a significant increase in the cellular Ca(2+) oscillation activity compared with RyR2 wild-type-expressing HEK293 cells. It was even more pronounced if glycine 1885 or 1886 was replaced by the acidic amino acids glutamate (G1885E) or aspartate (G1886D). Surprisingly, when both substitutions were introduced in the same RyR2 subunit (RyR2-G1885E/G1886S), the store-overload-induced calcium release activity was nearly completely abolished, although the Ca(2+) loading of the intracellular stores was markedly enhanced, and the channel still displayed substantial Ca(2+) release on stimulation by 5 mM caffeine. These results suggest that the adjacent glycines 1885 and 1886, located in the divergent region 3, are critical for the function and regulation of RyR2.
Collapse
|
35
|
Yamamoto T, Yano M, Xu X, Uchinoumi H, Tateishi H, Mochizuki M, Oda T, Kobayashi S, Ikemoto N, Matsuzaki M. Identification of target domains of the cardiac ryanodine receptor to correct channel disorder in failing hearts. Circulation 2008; 117:762-72. [PMID: 18227387 DOI: 10.1161/circulationaha.107.718957] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND We previously demonstrated that defective interdomain interaction between N-terminal (0 to 600) and central regions (2000 to 2500) of ryanodine receptor 2 (RyR2) induces Ca2+ leak in failing hearts and that K201 (JTV519) inhibits the Ca2+ leak by correcting the defective interdomain interaction. In the present report, we identified the K201-binding domain and characterized the role of this novel domain in the regulation of the RyR2 channel. METHODS AND RESULTS An assay using a quartz-crystal microbalance technique (a very sensitive mass-measuring technique) revealed that K201 specifically bound to recombinant RyR2 fragments 1741 to 2270 and 1981 to 2520 but not to other RyR2 fragments from the 1 to 2750 region (1 to 610, 494 to 1000, 741 to 1260, 985 to 1503, 1245 to 1768, 2234 to 2750). By further analysis of the fragment(1741-2270), K201 was found to specifically bind to its subfragment(2114-2149). With the use of the peptide matching this subfragment (DP(2114-2149)) as a carrier, the RyR2 was fluorescently labeled with methylcoumarin acetate (MCA) in a site-directed manner. After tryptic digestion, the major MCA-labeled fragment of RyR2 (155 kDa) was detected by an antibody raised against the central region (Ab(2132)). Moreover, of several recombinant RyR2 fragments, only fragment(2234-2750) was specifically MCA labeled; this suggests that the K201-binding domain(2114-2149) binds with domain(2234-2750). Addition of DP(2114-2149) to the MCA-labeled sarcoplasmic reticulum interfered with the interaction between domain(2114-2149) and domain(2234-2750), causing domain unzipping, as evidenced by an increased accessibility of the bound MCA to a large-size fluorescence quencher. In failing cardiomyocytes, the frequency of spontaneous Ca2+ spark was markedly increased compared with normal cardiomyocytes, whereas incorporation of DP(2114-2149) markedly decreased the frequency of spontaneous Ca2+ spark. CONCLUSIONS We first identified the K201-binding site as domain(2114-2149) of RyR2. Interruption of the interdomain interaction between the domain(2114-2149) and central domain(2234-2750) seems to mediate stabilization of RyR2 in failing hearts, which may lead to a novel therapeutic strategy against heart failure and perhaps lethal arrhythmia.
Collapse
Affiliation(s)
- Takeshi Yamamoto
- Department of Medicine and Clinical Science, Division of Cardiology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fleischer S. Personal recollections on the discovery of the ryanodine receptors of muscle. Biochem Biophys Res Commun 2008; 369:195-207. [PMID: 18182155 DOI: 10.1016/j.bbrc.2007.12.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 12/11/2007] [Indexed: 10/22/2022]
Abstract
The intracellular Ca(2+) release channels are indispensable molecular machinery in practically all eukaryotic cells of multicellular animals. They serve a key role in cell signaling by way of Ca(2+) as a second messenger. In response to a signaling event, the channels release Ca(2+) from intracellular stores. The resulting rise in cytoplasmic Ca(2+) concentration triggers the cell to carry out its specialized role, after which the intracellular Ca(2+) concentration must be reduced so that the signaling event can again be repeated. There are two types of intracellular Ca(2+) release channels, i.e., the ryanodine receptors and the inositol triphosphate receptors. My focus in this minireview is to present a personal account, from the vantage point our laboratory, of the discovery, isolation, and characterization of the ryanodine receptors from mammalian muscle. There are three isoforms: ryanodine receptor 1 (RyR1), first isolated from rabbit fast twitch skeletal muscle; ryanodine receptor 2 (RyR2), first isolated from dog heart; and ryanodine receptor 3 (RyR3), first isolated from bovine diaphragm muscle. The ryanodine receptors are the largest channel structures known. The RyR isoforms are very similar albeit with important differences. Natural mutations in humans in these receptors have already been associated with a number of muscle diseases.
Collapse
Affiliation(s)
- Sidney Fleischer
- Department of Biological Sciences, Vanderbilt University, 465 21st Avenue South, MRBIII Room 1210, Nashville, TN 37235, USA
| |
Collapse
|
37
|
Ryanodine receptor mutations in arrhythmias: advances in understanding the mechanisms of channel dysfunction. Biochem Soc Trans 2007; 35:946-51. [DOI: 10.1042/bst0350946] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The cardiac ryanodine receptor (RyR2) mediates rapid Ca2+ efflux from intracellular stores to effect myocyte contraction during the process of EC (excitation–contraction) coupling. It is now known that mutations in this channel perturb Ca2+ release function, leading to triggered arrhythmias that may cause SCD (sudden cardiac death). Resolving the precise molecular mechanisms by which SCD-linked RyR2 dysfunction occurs currently constitutes a burgeoning area of cardiac research. So far, defective channel phosphorylation, accessory protein binding, luminal/cytosolic Ca2+ sensing, and the disruption of interdomain interactions represent the main candidate mechanisms for explaining aberrant SR (sarcoplasmic reticulum) Ca2+ release via mutants of RyR2. It appears increasingly unlikely that a single exclusive common mechanism underlies every case of mutant channel dysfunction, and that each of these potential mechanisms may contribute to the resultant phenotype. The present review will consider very recent mechanistic developments in this field, including new observations from mutant RyR2 transgenic mouse models, peptide-probe studies, and the implications of functional and phenotypic heterogeneity of RyR2 mutations and polymorphisms.
Collapse
|
38
|
Bannister RA. Bridging the myoplasmic gap: recent developments in skeletal muscle excitation–contraction coupling. J Muscle Res Cell Motil 2007; 28:275-83. [PMID: 17899404 DOI: 10.1007/s10974-007-9118-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 08/28/2007] [Indexed: 01/17/2023]
Abstract
Conformational coupling between the L-type voltage-gated Ca(2+) channel (or 1,4-dihydropyridine receptor; DHPR) and the ryanodine-sensitive Ca(2+) release channel of the sarcoplasmic reticulum (RyR1) is the mechanistic basis for excitation-contraction (EC) coupling in skeletal muscle. In this article, recent findings regarding the roles of the individual cytoplasmic domains (the amino- and carboxyl-termini, cytoplasmic loops I-II, II-III, and III-IV) of the DHPR alpha(1S) subunit in bi-directional communication with RyR1 will be discussed.
Collapse
Affiliation(s)
- Roger A Bannister
- Department of Physiology and Biophysics, School of Medicine, University of Colorado at Denver and Health Sciences Center, RC-1, North Tower, P18-7130, Mail Stop F8307, 12800 E. 19th St, Aurora, CO 80045, USA.
| |
Collapse
|
39
|
Meng X, Xiao B, Cai S, Huang X, Li F, Bolstad J, Trujillo R, Airey J, Wayne Chen SR, Wagenknecht T, Liu Z. Three-dimensional localization of serine 2808, a phosphorylation site in cardiac ryanodine receptor. J Biol Chem 2007; 282:25929-39. [PMID: 17606610 PMCID: PMC2796423 DOI: 10.1074/jbc.m704474200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Type 2 ryanodine receptor (RyR2) is the major calcium release channel in cardiac muscle. Phosphorylation of RyR2 by cAMP-dependent protein kinase A and by calmodulin-dependent protein kinase II modulates channel activity. Hyperphosphorylation at a single amino acid residue, Ser-2808, has been proposed to directly disrupt the binding of a 12.6-kDa FK506-binding protein (FKBP12.6) to RyR2, causing a RyR2 malfunction that triggers cardiac arrhythmias in human heart failure. To determine the structural basis of the interaction between Ser-2808 and FKBP12.6, we have employed two independent approaches to map this phosphorylation site in RyR2 by three-dimensional cryo-electron microscopy. In one approach, we inserted a green fluorescent protein (GFP) after amino acid Tyr-2801, and mapped the GFP three-dimensional location in the RyR2 structure. In another approach, the binding site of monoclonal antibody 34C was mapped in the three-dimensional structure of skeletal muscle RyR1. The epitope of antibody 34C has been mapped to amino acid residues 2,756 through 2,803 of the RyR1 sequence, corresponding to residues 2,722 through 2,769 of the RyR2 sequence. These locations of GFP insertion and antibody binding are adjacent to one another in domain 6 of the cytoplasmic clamp region. Importantly, the three-dimensional location of the Ser-2808 phosphorylation site is 105-120 A distance from the FKBP12.6 binding site mapped previously, indicating that Ser-2808 is unlikely to be directly involved in the binding of FKBP12.6 to RyR2, as had been proposed previously.
Collapse
Affiliation(s)
- Xing Meng
- Wadsworth Center, New York State Department of Health, Albany, New York 12201
| | - Bailong Xiao
- Departments of Physiology and Biophysics, and of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Shitian Cai
- Departments of Physiology and Biophysics, and of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Xiaojun Huang
- Wadsworth Center, New York State Department of Health, Albany, New York 12201
- Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York 12201
| | - Fei Li
- Wadsworth Center, New York State Department of Health, Albany, New York 12201
- Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York 12201
| | - Jeff Bolstad
- Departments of Physiology and Biophysics, and of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Ramon Trujillo
- Wadsworth Center, New York State Department of Health, Albany, New York 12201
| | - Judith Airey
- Department of Pharmacology, University of Nevada, Reno, Nevada 89557
| | - S. R. Wayne Chen
- Departments of Physiology and Biophysics, and of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Terence Wagenknecht
- Wadsworth Center, New York State Department of Health, Albany, New York 12201
- Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, New York 12201
| | - Zheng Liu
- Wadsworth Center, New York State Department of Health, Albany, New York 12201
- To whom correspondence may be addressed: Wadsworth Center, New York State Department of Health, Albany, NY 12201. Tel.: 518-474-6516; Fax: 518-474-7992;
| |
Collapse
|
40
|
Wang R, Chen W, Cai S, Zhang J, Bolstad J, Wagenknecht T, Liu Z, Chen SRW. Localization of an NH(2)-terminal disease-causing mutation hot spot to the "clamp" region in the three-dimensional structure of the cardiac ryanodine receptor. J Biol Chem 2007; 282:17785-93. [PMID: 17452324 PMCID: PMC2800043 DOI: 10.1074/jbc.m700660200] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A region between residues 414 and 466 in the cardiac ryanodine receptor (RyR2) harbors more than half of the known NH(2)-terminal mutations associated with cardiac arrhythmias and sudden death. To gain insight into the structural basis of this NH(2)-terminal mutation hot spot, we have determined its location in the three-dimensional structure of RyR2. Green fluorescent protein (GFP), used as a structural marker, was inserted into the middle of this mutation hot spot after Ser-437 in the RyR2 sequence. The resultant GFP-RyR2 fusion protein, RyR2(S437-GFP,) was expressed in HEK293 cells and characterized using Ca(2+) release, [(3)H]ryanodine binding, and single cell Ca(2+) imaging studies. These functional analyses revealed that RyR2(S437-GFP) forms a caffeine- and ryanodine-sensitive Ca(2+) release channel that possesses Ca(2+) and caffeine dependence of activation indistinguishable from that of wild type (wt) RyR2. HEK293 cells expressing RyR2(S437-GFP) displayed a propensity for store overload-induced Ca(2+) release similar to that in cells expressing RyR2-wt. The three-dimensional structure of the purified RyR2(S437-GFP) was reconstructed using cryo-electron microscopy and single particle image processing. Subtraction of the three-dimensional reconstructions of RyR2-wt and RyR2(S437-GFP) revealed the location of the inserted GFP, and hence the NH(2)-terminal mutation hot spot, in a region between domains 5 and 9 in the clamp-shaped structure. This location is close to a previously mapped central disease-causing mutation site located in a region between domains 5 and 6. These results, together with findings from previous studies, suggest that the proposed interactions between the NH(2)-terminal and central regions of RyR2 are likely to take place between domains 5 and 6 and that the clamp-shaped structure, which shows substantial conformational differences between the closed and open states, is highly susceptible to disease-causing mutations.
Collapse
Affiliation(s)
- Ruiwu Wang
- Departments of Physiology & Biophysics, and of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Wenqian Chen
- Departments of Physiology & Biophysics, and of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Shitian Cai
- Departments of Physiology & Biophysics, and of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Jing Zhang
- Departments of Physiology & Biophysics, and of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Jeff Bolstad
- Departments of Physiology & Biophysics, and of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Terence Wagenknecht
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
- Department of Biomedical Sciences, School of Public Health, State University of New York at Albany, Albany, NY 12201
| | - Zheng Liu
- Wadsworth Center, New York State Department of Health, Albany, NY 12201
| | - S. R. Wayne Chen
- Departments of Physiology & Biophysics, and of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| |
Collapse
|
41
|
Abstract
Ryanodine receptors (RyRs)/Ca2+ release channels, on the endoplasmic and sarcoplasmic reticulum of most cell types, are required for intracellular Ca2+ release involved in diverse cellular functions, including muscle contraction and neurotransmitter release. The large cytoplasmic domain of the RyR serves as a scaffold for proteins that bind to and modulate the channel's function and that comprise a macromolecular signaling complex. These proteins include calstabins [FK506-binding proteins (FKBPs)], calmodulin (CaM), phosphodiesterase, kinases, phosphatases, and their cognate targeting proteins. This review focuses on recent progress in the understanding of RyR regulation and disease mechanisms that are associated with channel dysfunction.
Collapse
Affiliation(s)
- Ran Zalk
- Department of Physiology and Cellular Biophysics, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | | | |
Collapse
|
42
|
Serysheva II, Chiu W, Ludtke SJ. Single-particle electron cryomicroscopy of the ion channels in the excitation-contraction coupling junction. Methods Cell Biol 2007; 79:407-35. [PMID: 17327167 DOI: 10.1016/s0091-679x(06)79016-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Affiliation(s)
- Irina I Serysheva
- National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
43
|
Abstract
Triggered activity in cardiac muscle and intracellular Ca2+ have been linked in the past. However, today not only are there a number of cellular proteins that show clear Ca2+ dependence but also there are a number of arrhythmias whose mechanism appears to be linked to Ca2+-dependent processes. Thus we present a systematic review of the mechanisms of Ca2+ transport (forward excitation-contraction coupling) in the ventricular cell as well as what is known for other cardiac cell types. Second, we review the molecular nature of the proteins that are involved in this process as well as the functional consequences of both normal and abnormal Ca2+ cycling (e.g., Ca2+ waves). Finally, we review what we understand to be the role of Ca2+ cycling in various forms of arrhythmias, that is, those associated with inherited mutations and those that are acquired and resulting from reentrant excitation and/or abnormal impulse generation (e.g., triggered activity). Further solving the nature of these intricate and dynamic interactions promises to be an important area of research for a better recognition and understanding of the nature of Ca2+ and arrhythmias. Our solutions will provide a more complete understanding of the molecular basis for the targeted control of cellular calcium in the treatment and prevention of such.
Collapse
Affiliation(s)
- Henk E D J Ter Keurs
- Department of Medicine, Physiology and Biophysics, University of Calgary, Alberta, Canada
| | | |
Collapse
|
44
|
Ogawa Y. Dysregulation of the gain of CICR through ryanodine receptor1 (RyR1): the putative mechanism underlying malignant hyperthermia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 592:287-94. [PMID: 17278373 DOI: 10.1007/978-4-431-38453-3_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Yasuo Ogawa
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
45
|
Samsó M, Shen X, Allen PD. Structural Characterization of the RyR1–FKBP12 Interaction. J Mol Biol 2006; 356:917-27. [PMID: 16405911 DOI: 10.1016/j.jmb.2005.12.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Revised: 12/05/2005] [Accepted: 12/07/2005] [Indexed: 10/25/2022]
Abstract
The 12 kDa FK506-binding protein (FKBP12) constitutively binds to the calcium release channel RyR1. Removal of FKBP12 using FK506 or rapamycin causes an increased open probability and an increase in the frequency of sub-conductance states in RyR1. Using cryo-electron microscopy and single-particle image processing, we have determined the 3D difference map of FKBP12 associated with RyR1 at 16 A resolution that can be fitted with the atomic model of FKBP12 in a unique orientation. This has allowed us to better define the surfaces of close apposition between FKBP12 and RyR1. Our results shed light on the role of several FKBP12 residues that had been found critical for the specificity of the RyR1-FKBP12 interaction. As predicted from previous immunoprecipitation studies, our results suggest that Gln3 participates directly in this interaction. The orientation of RyR1-bound FKBP12, with part of its FK506 binding site facing towards RyR1, allows us to propose how FK506 is involved in the dissociation of FKBP12 from RyR1.
Collapse
Affiliation(s)
- Montserrat Samsó
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
46
|
George CH, Jundi H, Walters N, Thomas NL, West RR, Lai FA. Arrhythmogenic Mutation-Linked Defects in Ryanodine Receptor Autoregulation Reveal a Novel Mechanism of Ca
2+
Release Channel Dysfunction. Circ Res 2006; 98:88-97. [PMID: 16339485 DOI: 10.1161/01.res.0000199296.70534.7c] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Arrhythmogenic cardiac ryanodine receptor (RyR2) mutations are associated with stress-induced malignant tachycardia, frequently leading to sudden cardiac death (SCD). The causative mechanisms of RyR2 Ca
2+
release dysregulation are complex and remain controversial. We investigated the functional impact of clinically-severe RyR2 mutations occurring in the central domain, and the C-terminal I domain, a key locus of RyR2 autoregulation, on interdomain interactions and Ca
2+
release in living cells. Using high-resolution confocal microscopy and fluorescence resonance energy transfer (FRET) analysis of interaction between fusion proteins corresponding to amino- (N-) and carboxyl- (C-) terminal RyR2 domains, we determined that in resting cells, RyR2 interdomain interaction remained unaltered after introduction of SCD-linked mutations and normal Ca
2+
regulation was maintained. In contrast, after channel activation, the abnormal Ca
2+
release via mutant RyR2 was intrinsically linked to altered interdomain interaction that was equivalent with all mutations and exhibited threshold characteristics (caffeine >2.5 mmol/L; Ca
2+
>150 nmol/L). Noise analysis revealed that I domain mutations introduced a distinct pattern of conformational instability in Ca
2+
handling and interdomain interaction after channel activation that was absent in signals obtained from the central domain mutation. I domain–linked channel instability also occurred in intact RyR2 expressed in CHO cells and in HL-1 cardiomyocytes. These new insights highlight a critical role for mutation-linked defects in channel autoregulation, and may contribute to a molecular explanation for the augmented Ca
2+
release following RyR2 channel activation. Our findings also suggest that the mutational locus may be an important mechanistic determinant of Ca
2+
release channel dysfunction in arrhythmia and SCD.
Collapse
Affiliation(s)
- Christopher H George
- Department of Cardiology, Wales Heart Research Institute, Cardiff University School of Medicine, Cardiff, UK.
| | | | | | | | | | | |
Collapse
|