1
|
Tourel A, Reynaud-Dulaurier R, Brocard J, Fauré J, Marty I, Petiot A. RyR1 Is Involved in the Control of Myogenesis. Cells 2025; 14:158. [PMID: 39936950 PMCID: PMC11817019 DOI: 10.3390/cells14030158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
The RyR1 calcium release channel is a key player in skeletal muscle excitation-contraction coupling. Mutations in the RYR1 gene are associated with congenital myopathies. Recently, a role of RyR1 in myotubes differentiation has been proposed and attributed to its calcium channel function, which nonetheless remains to be clearly demonstrated. In order to clarify RyR1 role in myogenesis, we have developed an in vitro model, the so-called RyR1-Rec myotubes, which are mouse primary myotubes with an inducible decrease in RyR1 protein amount and in RyR1-mediated calcium release. Using this model, we showed that the RyR1 protein decrease was responsible for an increase in both differentiation and fusion, from the RNA level to the morphological level, without affecting the myogenic factors MyoD and MyoG. Although an increase in mTOR pathway was observed in RyR1-Rec myotubes, it did not seem to be responsible for the role of RyR1 in myogenesis. Additionally, even if modulation of intracellular calcium level affected RyR1-Rec myotubes differentiation, we have shown that the role of RyR1 in myogenesis was independent of its calcium channel function. Therefore, our findings indicate that, besides its pivotal role as a calcium channel responsible for muscle contraction, RyR1 fulfills a calcium-independent inhibitor function of myogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Isabelle Marty
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, 38000 Grenoble, France (R.R.-D.); (J.B.); (J.F.)
| | - Anne Petiot
- University Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, 38000 Grenoble, France (R.R.-D.); (J.B.); (J.F.)
| |
Collapse
|
2
|
Beaufils M, Melka M, Brocard J, Benoit C, Debbah N, Mamchaoui K, Romero NB, Dalmas-Laurent AF, Quijano-Roy S, Fauré J, Rendu J, Marty I. Functional benefit of CRISPR-Cas9-induced allele deletion for RYR1 dominant mutation. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102259. [PMID: 39071953 PMCID: PMC11278293 DOI: 10.1016/j.omtn.2024.102259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/14/2024] [Indexed: 07/30/2024]
Abstract
More than 700 pathogenic or probably pathogenic variations have been identified in the RYR1 gene causing various myopathies collectively known as "RYR1-related myopathies." There is no treatment for these myopathies, and gene therapy stands out as one of the most promising approaches. In the context of a dominant form of central core disease due to a RYR1 mutation, we aimed at showing the functional benefit of inactivating specifically the mutated RYR1 allele by guiding CRISPR-Cas9 cleavages onto frequent single-nucleotide polymorphisms (SNPs) segregating on the same chromosome. Whole-genome sequencing was used to pinpoint SNPs localized on the mutant RYR1 allele and identified specific CRISPR-Cas9 guide RNAs. Lentiviruses encoding these guide RNAs and the SpCas9 nuclease were used to transduce immortalized patient myoblasts, inducing the specific deletion of the mutant RYR1 allele. The efficiency of the deletion was assessed at DNA and RNA levels, and at the functional level after monitoring calcium release induced by the stimulation of the RyR1-channel. This study provides in cellulo proof of concept regarding the benefits of mutant RYR1 allele deletion, in the case of a dominant RYR1 mutation, from both a molecular and functional perspective, and could apply potentially to 20% of all patients with a RYR1 mutation.
Collapse
Affiliation(s)
- Mathilde Beaufils
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Margaux Melka
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julie Brocard
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Clement Benoit
- University Grenoble Alpes, TIMC, CNRS UMR5525, 38000 Grenoble, France
| | - Nagi Debbah
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
- University Grenoble Alpes, TIMC, CNRS UMR5525, 38000 Grenoble, France
- University Grenoble Alpes, Departement de Pharmacochimie Moléculaire, CNRS UMR 5063, 38400 Saint-Martin-d'Hères, France
| | - Kamel Mamchaoui
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75000 Paris, France
| | - Norma B. Romero
- Neuromuscular Morphology Unit, Institut de Myologie, Pitié-Salpêtrière Hospital, Sorbonne Université, 75000 Paris, France
| | | | - Susana Quijano-Roy
- Neuromuscular Unit (NEIDF), Child Neurology and ICU Department, Raymond-Poincaré Hospital, (APHP University Paris-Saclay), 92380 Garches, France
| | - Julien Fauré
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - John Rendu
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Isabelle Marty
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| |
Collapse
|
3
|
Synthesis of Dihydropyrimidines: Isosteres of Nifedipine and Evaluation of Their Calcium Channel Blocking Efficiency. Molecules 2023; 28:molecules28020784. [PMID: 36677842 PMCID: PMC9867414 DOI: 10.3390/molecules28020784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023] Open
Abstract
Hypertension and cardiovascular diseases related to it remain the leading medical challenges globally. Several drugs have been synthesized and commercialized to manage hypertension. Some of these drugs have a dihydropyrimidine skeleton structure, act as efficient calcium channel blockers, and affect the calcium ions' intake in vascular smooth muscle, hence managing hypertension. The synthesis of such moieties is crucial, and documenting their structure-activity relationship, their evolved and advanced synthetic procedures, and future opportunities in this area is currently a priority. Tremendous efforts have been made after the discovery of the Biginelli condensation reaction in the synthesis of dihydropyrimidines. From the specific selection of Biginelli adducts to the variation in the formed intermediates to achieve target compounds containing heterocylic rings, aldehydes, a variety of ketones, halogens, and many other desired functionalities, extensive studies have been carried out. Several substitutions at the C3, C4, and C5 positions of dihydropyrimidines have been explored, aiming to produce feasible derivatives with acceptable yields as well as antihypertensive activity. The current review aims to cover this requirement in detail.
Collapse
|
4
|
Chivet M, McCluskey M, Nicot AS, Brocard J, Beaufils M, Giovannini D, Giannesini B, Poreau B, Brocard J, Humbert S, Saudou F, Fauré J, Marty I. Huntingtin regulates calcium fluxes in skeletal muscle. J Gen Physiol 2022; 155:213700. [PMID: 36409218 PMCID: PMC9682417 DOI: 10.1085/jgp.202213103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/09/2022] [Accepted: 09/03/2022] [Indexed: 11/22/2022] Open
Abstract
The expression of the Huntingtin protein, well known for its involvement in the neurodegenerative Huntington's disease, has been confirmed in skeletal muscle. The impact of HTT deficiency was studied in human skeletal muscle cell lines and in a mouse model with inducible and muscle-specific HTT deletion. Characterization of calcium fluxes in the knock-out cell lines demonstrated a reduction in excitation-contraction (EC) coupling, related to an alteration in the coupling between the dihydropyridine receptor and the ryanodine receptor, and an increase in the amount of calcium stored within the sarcoplasmic reticulum, linked to the hyperactivity of store-operated calcium entry (SOCE). Immunoprecipitation experiments demonstrated an association of HTT with junctophilin 1 (JPH1) and stromal interaction molecule 1 (STIM1), both providing clues on the functional effects of HTT deletion on calcium fluxes. Characterization of muscle strength and muscle anatomy of the muscle-specific HTT-KO mice demonstrated that HTT deletion induced moderate muscle weakness and mild muscle atrophy associated with histological abnormalities, similar to the phenotype observed in tubular aggregate myopathy. Altogether, this study points toward the hypotheses of the involvement of HTT in EC coupling via its interaction with JPH1, and on SOCE via its interaction with JPH1 and/or STIM1.
Collapse
Affiliation(s)
- Mathilde Chivet
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Maximilian McCluskey
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Anne Sophie Nicot
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Julie Brocard
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Mathilde Beaufils
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Diane Giovannini
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Benoit Giannesini
- Centre National de la Recherche Scientifique, Centre de Résonance Magnétique Biologique et Médicale, Aix Marseille University, Marseille, France
| | - Brice Poreau
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Jacques Brocard
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Sandrine Humbert
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Frédéric Saudou
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Julien Fauré
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France
| | - Isabelle Marty
- CHU Grenoble Alpes, Grenoble Institut Neurosciences, INSERM, U1216, Université Grenoble Alpes, Grenoble, France,Correspondence to Isabelle Marty:
| |
Collapse
|
5
|
Heck J, Palmeira Do Amaral AC, Weißbach S, El Khallouqi A, Bikbaev A, Heine M. More than a pore: How voltage-gated calcium channels act on different levels of neuronal communication regulation. Channels (Austin) 2021; 15:322-338. [PMID: 34107849 PMCID: PMC8205089 DOI: 10.1080/19336950.2021.1900024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) represent key regulators of the calcium influx through the plasma membrane of excitable cells, like neurons. Activated by the depolarization of the membrane, the opening of VGCCs induces very transient and local changes in the intracellular calcium concentration, known as calcium nanodomains, that in turn trigger calcium-dependent signaling cascades and the release of chemical neurotransmitters. Based on their central importance as concierges of excitation-secretion coupling and therefore neuronal communication, VGCCs have been studied in multiple aspects of neuronal function and malfunction. However, studies on molecular interaction partners and recent progress in omics technologies have extended the actual concept of these molecules. With this review, we want to illustrate some new perspectives of VGCCs reaching beyond their function as calcium-permeable pores in the plasma membrane. Therefore, we will discuss the relevance of VGCCs as voltage sensors in functional complexes with ryanodine receptors, channel-independent actions of auxiliary VGCC subunits, and provide an insight into how VGCCs even directly participate in gene regulation. Furthermore, we will illustrate how structural changes in the intracellular C-terminus of VGCCs generated by alternative splicing events might not only affect the biophysical channel characteristics but rather determine their molecular environment and downstream signaling pathways.
Collapse
Affiliation(s)
- Jennifer Heck
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Ana Carolina Palmeira Do Amaral
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Stephan Weißbach
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
- Computational Genomics and Bioinformatics, Johannes Gutenberg-University Mainz, University Medical Center Mainz, Institute for Human Genetics, Mainz, Germany
| | - Abderazzaq El Khallouqi
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Arthur Bikbaev
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Martin Heine
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| |
Collapse
|
6
|
Pelletier L, Petiot A, Brocard J, Giannesini B, Giovannini D, Sanchez C, Travard L, Chivet M, Beaufils M, Kutchukian C, Bendahan D, Metzger D, Franzini Armstrong C, Romero NB, Rendu J, Jacquemond V, Fauré J, Marty I. In vivo RyR1 reduction in muscle triggers a core-like myopathy. Acta Neuropathol Commun 2020; 8:192. [PMID: 33176865 PMCID: PMC7657350 DOI: 10.1186/s40478-020-01068-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
Mutations in the RYR1 gene, encoding the skeletal muscle calcium channel RyR1, lead to congenital myopathies, through expression of a channel with abnormal permeability and/or in reduced amount, but the direct functional whole organism consequences of exclusive reduction in RyR1 amount have never been studied. We have developed and characterized a mouse model with inducible muscle specific RYR1 deletion. Tamoxifen-induced recombination in the RYR1 gene at adult age resulted in a progressive reduction in the protein amount reaching a stable level of 50% of the initial amount, and was associated with a progressive muscle weakness and atrophy. Measurement of calcium fluxes in isolated muscle fibers demonstrated a reduction in the amplitude of RyR1-related calcium release mirroring the reduction in the protein amount. Alterations in the muscle structure were observed, with fibers atrophy, abnormal mitochondria distribution and membrane remodeling. An increase in the expression level of many proteins was observed, as well as an inhibition of the autophagy process. This model demonstrates that RyR1 reduction is sufficient to recapitulate most features of Central Core Disease, and accordingly similar alterations were observed in muscle biopsies from Dusty Core Disease patients (a subtype of Central Core Disease), pointing to common pathophysiological mechanisms related to RyR1 reduction.
Collapse
|
7
|
Sébastien M, Aubin P, Brocard J, Brocard J, Marty I, Fauré J. Dynamics of triadin, a muscle-specific triad protein, within sarcoplasmic reticulum subdomains. Mol Biol Cell 2020; 31:261-272. [PMID: 31877066 PMCID: PMC7183767 DOI: 10.1091/mbc.e19-07-0399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In skeletal muscle, proteins of the calcium release complex responsible for the excitation-contraction (EC) coupling are exclusively localized in specific reticulum–plasma membrane (ER-PM) contact points named triads. The CRC protein triadin (T95) is localized in the sarcoplasmic reticulum (SR) subdomain of triads where it forms large multimers. However, the mechanisms leading to the steady-state accumulation of T95 in these specific areas of SR are largely unknown. To visualize T95 dynamics, fluorescent chimeras were expressed in triadin knockout myotubes, and their mobility was compared with the mobility of Sec61β, a membrane protein of the SR unrelated to the EC coupling process. At all stages of skeletal muscle cells differentiation, we show a permanent flux of T95 diffusing in the SR membrane. Moreover, we find evidence that a longer residence time in the ER-PM contact point is due to the transmembrane domain of T95 resulting in an overall triad localization.
Collapse
Affiliation(s)
- Muriel Sébastien
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France
| | - Perrine Aubin
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France
| | - Jacques Brocard
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France
| | - Julie Brocard
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France
| | - Isabelle Marty
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France
| | - Julien Fauré
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France.,Grenoble Institut Neurosciences, Inserm, U1216, CHU Grenoble Alpes, University Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
8
|
Fodor J, Al-Gaadi D, Czirják T, Oláh T, Dienes B, Csernoch L, Szentesi P. Improved Calcium Homeostasis and Force by Selenium Treatment and Training in Aged Mouse Skeletal Muscle. Sci Rep 2020; 10:1707. [PMID: 32015413 PMCID: PMC6997352 DOI: 10.1038/s41598-020-58500-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
During aging reduction in muscle mass (sarcopenia) and decrease in physical activity lead to partial loss of muscle force and increased fatigability. Deficiency in the essential trace element selenium might augment these symptoms as it can cause muscle pain, fatigue, and proximal weakness. Average voluntary daily running, maximal twitch and tetanic force, and calcium release from the sarcoplasmic reticulum (SR) decreased while reactive oxygen species (ROS) production associated with tetanic contractions increased in aged – 22-month-old – as compared to young – 4-month-old – mice. These changes were accompanied by a decline in the ryanodine receptor type 1 (RyR1) and Selenoprotein N content and the increased amount of a degraded RyR1. Both lifelong training and selenium supplementation, but not the presence of an increased muscle mass at young age, were able to compensate for the reduction in muscle force and SR calcium release with age. Selenium supplementation was also able to significantly enhance the Selenoprotein N levels in aged mice. Our results describe, for the first time, the beneficial effects of selenium supplementation on calcium release from the SR and muscle force in old age while point out that increased muscle mass does not improve physical performance with aging.
Collapse
Affiliation(s)
- János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dána Al-Gaadi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Czirják
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
9
|
Cacheux M, Fauconnier J, Thireau J, Osseni A, Brocard J, Roux-Buisson N, Brocard J, Fauré J, Lacampagne A, Marty I. Interplay between Triadin and Calsequestrin in the Pathogenesis of CPVT in the Mouse. Mol Ther 2019; 28:171-179. [PMID: 31607542 DOI: 10.1016/j.ymthe.2019.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 01/07/2023] Open
Abstract
Recessive forms of catecholaminergic polymorphic ventricular tachycardia (CPVT) are induced by mutations in genes encoding triadin or calsequestrin, two proteins that belong to the Ca2+ release complex, responsible for intracellular Ca2+ release triggering cardiac contractions. To better understand the mechanisms of triadin-induced CPVT and to assay multiple therapeutic interventions, we used a triadin knockout mouse model presenting a CPVT-like phenotype associated with a decrease in calsequestrin protein level. We assessed different approaches to rescue protein expression and to correct intracellular Ca2+ release and cardiac function: pharmacological treatment with kifunensine or a viral gene transfer-based approach, using adeno-associated virus serotype 2/9 (AAV2/9) encoding the triadin or calsequestrin. We observed that the levels of triadin and calsequestrin are intimately linked, and that reduction of both proteins contributes to the CPVT phenotype. Different combinations of triadin and calsequestrin expression level were obtained using these therapeutic approaches. A full expression of each is not necessary to correct the phenotype; a fine-tuning of the relative re-expression of both triadin and calsequestrin is required to correct the CPVT phenotype and rescue the cardiac function. AAV-mediated gene delivery of calsequestrin or triadin and treatment with kifunensine are potential treatments for recessive forms of CPVT due to triadin mutations.
Collapse
Affiliation(s)
- Marine Cacheux
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Jérémy Fauconnier
- University of Montpellier, INSERM U1046, CNRS 9214, CHU Montpellier, 34295 Montpellier, France
| | - Jérôme Thireau
- University of Montpellier, INSERM U1046, CNRS 9214, CHU Montpellier, 34295 Montpellier, France
| | - Alexis Osseni
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Jacques Brocard
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Nathalie Roux-Buisson
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Julie Brocard
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Julien Fauré
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Alain Lacampagne
- University of Montpellier, INSERM U1046, CNRS 9214, CHU Montpellier, 34295 Montpellier, France.
| | - Isabelle Marty
- Grenoble Institut Neurosciences, INSERM, Grenoble Alpes University, U1216, CHU Grenoble Alpes, 38700 La Tronche, France.
| |
Collapse
|
10
|
Saini J, Faroni A, Abd Al Samid M, Reid AJ, Lightfoot AP, Mamchaoui K, Mouly V, Butler-Browne G, McPhee JS, Degens H, Al-Shanti N. Simplified in vitro engineering of neuromuscular junctions between rat embryonic motoneurons and immortalized human skeletal muscle cells. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2019; 12:1-9. [PMID: 30863121 PMCID: PMC6388735 DOI: 10.2147/sccaa.s187655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background Neuromuscular junctions (NMJs) consist of the presynaptic cholinergic motoneuron terminals and the corresponding postsynaptic motor endplates on skeletal muscle fibers. At the NMJ the action potential of the neuron leads, via release of acetylcholine, to muscle membrane depolarization that in turn is translated into muscle contraction and physical movement. Despite the fact that substantial NMJ research has been performed, the potential of in vivo NMJ investigations is inadequate and difficult to employ. A simple and reproducible in vitro NMJ model may provide a robust means to study the impact of neurotrophic factors, growth factors, and hormones on NMJ formation, structure, and function. Methods This report characterizes a novel in vitro NMJ model utilizing immortalized human skeletal muscle stem cells seeded on 35 mm glass-bottom dishes, cocultured and innervated with spinal cord explants from rat embryos at ED 13.5. The cocultures were fixed and stained on day 14 for analysis and assessment of NMJ formation and development. Results This unique serum- and trophic factor-free system permits the growth of cholinergic motoneurons, the formation of mature NMJs, and the development of highly differentiated contractile myotubes, which exhibit appropriate configuration of transversal triads, representative of in vivo conditions. Conclusion This coculture system provides a tool to study vital features of NMJ formation, regulation, maintenance, and repair, as well as a model platform to explore neuromuscular diseases and disorders affecting NMJs.
Collapse
Affiliation(s)
- Jasdeep Saini
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Department of Plastic Surgery & Burns, University Hospitals of South Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Marwah Abd Al Samid
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Adam J Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Department of Plastic Surgery & Burns, University Hospitals of South Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Adam P Lightfoot
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Kamel Mamchaoui
- Center for Research in Myology, Sorbonne Université- INSERM, Paris, France
| | - Vincent Mouly
- Center for Research in Myology, Sorbonne Université- INSERM, Paris, France
| | | | - Jamie S McPhee
- Department of Sport and Exercise Science, Manchester Metropolitan University, Manchester, UK
| | - Hans Degens
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK, .,Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania.,University of Medicine and Pharmacy of Targu Mures, Targu Mures, Romania
| | - Nasser Al-Shanti
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| |
Collapse
|
11
|
Garibaldi M, Rendu J, Brocard J, Lacene E, Fauré J, Brochier G, Beuvin M, Labasse C, Madelaine A, Malfatti E, Bevilacqua JA, Lubieniecki F, Monges S, Taratuto AL, Laporte J, Marty I, Antonini G, Romero NB. 'Dusty core disease' (DuCD): expanding morphological spectrum of RYR1 recessive myopathies. Acta Neuropathol Commun 2019; 7:3. [PMID: 30611313 PMCID: PMC6320585 DOI: 10.1186/s40478-018-0655-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Several morphological phenotypes have been associated to RYR1-recessive myopathies. We recharacterized the RYR1-recessive morphological spectrum by a large monocentric study performed on 54 muscle biopsies from a large cohort of 48 genetically confirmed patients, using histoenzymology, immunohistochemistry, and ultrastructural studies. We also analysed the level of RyR1 expression in patients’ muscle biopsies. We defined “dusty cores” the irregular areas of myofibrillar disorganisation characterised by a reddish-purple granular material deposition with uneven oxidative stain and devoid of ATPase activity, which represent the characteristic lesion in muscle biopsy in 54% of patients. We named Dusty Core Disease (DuCD) the corresponding entity of congenital myopathy. Dusty cores had peculiar histological and ultrastructural characteristics compared to the other core diseases. DuCD muscle biopsies also showed nuclear centralization and type1 fibre predominance. Dusty cores were not observed in other core myopathies and centronuclear myopathies. The other morphological groups in our cohort of patients were: Central Core (CCD: 21%), Core-Rod (C&R:15%) and Type1 predominance “plus” (T1P+:10%). DuCD group was associated to an earlier disease onset, a more severe clinical phenotype and a lowest level of RyR1 expression in muscle, compared to the other groups. Variants located in the bridge solenoid and the pore domains were more frequent in DuCD patients. In conclusion, DuCD is the most frequent histopathological presentation of RYR1-recessive myopathies. Dusty cores represent the unifying morphological lesion among the DuCD pathology spectrum and are the morphological hallmark for the recessive form of disease.
Collapse
|
12
|
Sébastien M, Giannesini B, Aubin P, Brocard J, Chivet M, Pietrangelo L, Boncompagni S, Bosc C, Brocard J, Rendu J, Gory-Fauré S, Andrieux A, Fourest-Lieuvin A, Fauré J, Marty I. Deletion of the microtubule-associated protein 6 (MAP6) results in skeletal muscle dysfunction. Skelet Muscle 2018; 8:30. [PMID: 30231928 PMCID: PMC6147105 DOI: 10.1186/s13395-018-0176-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/12/2018] [Indexed: 01/23/2023] Open
Abstract
Background The skeletal muscle fiber has a specific and precise intracellular organization which is at the basis of an efficient muscle contraction. Microtubules are long known to play a major role in the function and organization of many cells, but in skeletal muscle, the contribution of the microtubule cytoskeleton to the efficiency of contraction has only recently been studied. The microtubule network is dynamic and is regulated by many microtubule-associated proteins (MAPs). In the present study, the role of the MAP6 protein in skeletal muscle organization and function has been studied using the MAP6 knockout mouse line. Methods The presence of MAP6 transcripts and proteins was shown in mouse muscle homogenates and primary culture using RT-PCR and western blot. The in vivo evaluation of muscle force of MAP6 knockout (KO) mice was performed on anesthetized animals using electrostimulation coupled to mechanical measurement and multimodal magnetic resonance. The impact of MAP6 deletion on microtubule organization and intracellular structures was studied using immunofluorescent labeling and electron microscopy, and on calcium release for muscle contraction using Fluo-4 calcium imaging on cultured myotubes. Statistical analysis was performed using Student’s t test or the Mann-Whitney test. Results We demonstrate the presence of MAP6 transcripts and proteins in skeletal muscle. Deletion of MAP6 results in a large number of muscle modifications: muscle weakness associated with slight muscle atrophy, alterations of microtubule network and sarcoplasmic reticulum organization, and reduction in calcium release. Conclusion Altogether, our results demonstrate that MAP6 is involved in skeletal muscle function. Its deletion results in alterations in skeletal muscle contraction which contribute to the global deleterious phenotype of the MAP6 KO mice. As MAP6 KO mouse line is a model for schizophrenia, our work points to a possible muscle weakness associated to some forms of schizophrenia. Electronic supplementary material The online version of this article (10.1186/s13395-018-0176-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Muriel Sébastien
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | | | - Perrine Aubin
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Julie Brocard
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Mathilde Chivet
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Laura Pietrangelo
- CeSI-Met & DNICS, University G. d' Annunzio of Chieti, I-66100, Chieti, Italy
| | - Simona Boncompagni
- CeSI-Met & DNICS, University G. d' Annunzio of Chieti, I-66100, Chieti, Italy
| | - Christophe Bosc
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Jacques Brocard
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - John Rendu
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CHU Grenoble, Biochimie et Génétique Moléculaire, F-38000, Grenoble, France
| | - Sylvie Gory-Fauré
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France
| | - Annie Andrieux
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CEA-Grenoble, BIG, F-38000, Grenoble, France
| | - Anne Fourest-Lieuvin
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CEA-Grenoble, BIG, F-38000, Grenoble, France
| | - Julien Fauré
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France.,University Grenoble Alpes, F-38000, Grenoble, France.,CHU Grenoble, Biochimie et Génétique Moléculaire, F-38000, Grenoble, France
| | - Isabelle Marty
- INSERM 1216, Grenoble Institute of Neurosciences, F-38000, Grenoble, France. .,University Grenoble Alpes, F-38000, Grenoble, France. .,GIN- Inserm U1216 - Bat EJ Safra, Chemin Fortuné Ferrini, 38700, La Tronche, France.
| |
Collapse
|
13
|
Pritchard HAT, Gonzales AL, Pires PW, Drumm BT, Ko EA, Sanders KM, Hennig GW, Earley S. Microtubule structures underlying the sarcoplasmic reticulum support peripheral coupling sites to regulate smooth muscle contractility. Sci Signal 2017; 10:eaan2694. [PMID: 28928237 PMCID: PMC6328376 DOI: 10.1126/scisignal.aan2694] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Junctional membrane complexes facilitate excitation-contraction coupling in skeletal and cardiac muscle cells by forming subcellular invaginations that maintain close (≤20 nm) proximity of ryanodine receptors (RyRs) on the sarcoplasmic reticulum (SR) with voltage-dependent Ca2+ channels in the plasma membrane. In fully differentiated smooth muscle cells, junctional membrane complexes occur as distributed sites of peripheral coupling. We investigated the role of the cytoskeleton in maintaining peripheral coupling and associated Ca2+ signaling networks within native smooth muscle cells of mouse and rat cerebral arteries. Using live-cell confocal and superresolution microscopy, we found that the tight interactions between the SR and the plasma membrane in these cells relied on arching microtubule structures present at the periphery of smooth muscle cells and were independent of the actin cytoskeleton. Loss of peripheral coupling associated with microtubule depolymerization altered the spatiotemporal properties of localized Ca2+ sparks generated by the release of Ca2+ through type 2 RyRs (RyR2s) on the SR and decreased the number of sites of colocalization between RyR2s and large-conductance Ca2+-activated K+ (BK) channels. The reduced BK channel activity associated with the loss of SR-plasma membrane interactions was accompanied by increased pressure-induced constriction of cerebral resistance arteries. We conclude that microtubule structures maintain peripheral coupling in contractile smooth muscle cells, which is crucial for the regulation of contractility and cerebral vascular tone.
Collapse
Affiliation(s)
- Harry A T Pritchard
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Albert L Gonzales
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, USA
| | - Paulo W Pires
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Eun A Ko
- Department of Physiology and Cell Biology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Grant W Hennig
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, USA
| | - Scott Earley
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA.
| |
Collapse
|
14
|
Osseni A, Sébastien M, Sarrault O, Baudet M, Couté Y, Fauré J, Fourest-Lieuvin A, Marty I. Triadin and CLIMP-63 form a link between triads and microtubules in muscle cells. J Cell Sci 2016; 129:3744-3755. [PMID: 27562070 DOI: 10.1242/jcs.188862] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/18/2016] [Indexed: 01/20/2023] Open
Abstract
In skeletal muscle, the triad is a structure comprising a transverse (T)-tubule and sarcoplasmic reticulum (SR) cisternae. Triads constitute the basis of excitation-contraction coupling as the cradle of the Ca2+ release complex. We have shown previously that triadin, a member of this complex, has shaping properties on reticulum membrane and is indirectly involved in a link between triads and microtubules. We have identified here that CLIMP-63 (also known as CKAP4), as the partner of triadin, is responsible for this association of triads and microtubules. Triadin and CLIMP-63 interact through their respective luminal domains and the shaping properties of triadin depend on the capacity of CLIMP-63 to bind microtubules with its cytosolic portion. In skeletal muscle, CLIMP-63 is localized in the SR, including triads, and is associated with the Ca2+ release complex through its interaction with triadin. Knockout of triadin in muscles results in the delocalization of CLIMP-63 from triads, its dissociation from the Ca2+ release complex and a disorganization of the microtubule network. Our results suggest that the association of triadin and CLIMP-63 could be involved in the shaping of SR terminal cisternae and in the guidance of microtubules close to the triads.
Collapse
Affiliation(s)
- Alexis Osseni
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble F-38000, France INSERM, U1216, Grenoble F-38000, France
| | - Muriel Sébastien
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble F-38000, France INSERM, U1216, Grenoble F-38000, France
| | - Oriana Sarrault
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble F-38000, France INSERM, U1216, Grenoble F-38000, France
| | - Mathieu Baudet
- University Grenoble Alpes, BIG-BGE, Grenoble F-38000, France CEA, BIG, BGE and GPC, Grenoble F-38000, France INSERM, BGE, Grenoble F-38000, France
| | - Yohann Couté
- University Grenoble Alpes, BIG-BGE, Grenoble F-38000, France CEA, BIG, BGE and GPC, Grenoble F-38000, France INSERM, BGE, Grenoble F-38000, France
| | - Julien Fauré
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble F-38000, France INSERM, U1216, Grenoble F-38000, France CHU de Grenoble, Grenoble F-38000, France
| | - Anne Fourest-Lieuvin
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble F-38000, France INSERM, U1216, Grenoble F-38000, France CEA, BIG, BGE and GPC, Grenoble F-38000, France
| | - Isabelle Marty
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble F-38000, France INSERM, U1216, Grenoble F-38000, France
| |
Collapse
|
15
|
Cacheux M, Blum A, Sébastien M, Wozny AS, Brocard J, Mamchaoui K, Mouly V, Roux-Buisson N, Rendu J, Monnier N, Krivosic R, Allen P, Lacour A, Lunardi J, Fauré J, Marty I. Functional Characterization of a Central Core Disease RyR1 Mutation (p.Y4864H) Associated with Quantitative Defect in RyR1 Protein. J Neuromuscul Dis 2015; 2:421-432. [PMID: 27858745 PMCID: PMC5240544 DOI: 10.3233/jnd-150073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Central Core Disease (CCD) is a congenital myopathy often resulting from a mutation in RYR1 gene. Mutations in RyR1 can increase or decrease channel activity, or induce a reduction in the amount of protein. The consequences of a single mutation are sometimes multiple and the analysis of the functional effects is complex. Objective: The consequences of the p.Y4864H mutation identified in a CCD patient have been studied regarding both RyR1 function and amount. Methods: The amount of RyR1 in human and mouse muscles was evaluated using qRT-PCR and quantitative Western blot, and calcium release was studied using calcium imaging on primary cultures. The results were compared between human and mouse. Results: The p.Y4864H mutation induced an alteration of calcium release, and in addition was associated to a reduction in the amount of RyR1 in the patient’s muscle. This suggests two possible pathophysiological mechanisms: the alteration of calcium release could result from a modification of the channel properties of RyR1 or from a RyR1 reduction. In order to discriminate between the two hypotheses, we used the heterozygous RyR1 knockout (RyR1+/–) mouse model showing a comparable RyR1 protein reduction. No reduction in calcium release was observed in primary muscle culture from these mice, and no muscle weakness was measured. Conclusions: Because the reduction in the amount of RyR1 protein has no functional consequences in the murine model, the muscle weakness observed in the patient is most likely the result of a modification of the calcium channel function of RyR1 due to the p.Y4864H mutation.
Collapse
Affiliation(s)
- Marine Cacheux
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France
| | - Ariane Blum
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France
| | - Muriel Sébastien
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France
| | - Anne Sophie Wozny
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France.,Centre Hospitalier Régional Universitaire de Grenoble, Hôpital Michallon, Biochimie Génétique et Moléculaire, Grenoble, France
| | - Julie Brocard
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France
| | - Kamel Mamchaoui
- UMRS974 Inserm, UMR7215 CNRS, Institut de Myologie, GH PitiéSalpétrière, 47 bd de l'hôpital, Paris, France
| | - Vincent Mouly
- UMRS974 Inserm, UMR7215 CNRS, Institut de Myologie, GH PitiéSalpétrière, 47 bd de l'hôpital, Paris, France
| | - Nathalie Roux-Buisson
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France.,Centre Hospitalier Régional Universitaire de Grenoble, Hôpital Michallon, Biochimie Génétique et Moléculaire, Grenoble, France
| | - John Rendu
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France.,Centre Hospitalier Régional Universitaire de Grenoble, Hôpital Michallon, Biochimie Génétique et Moléculaire, Grenoble, France
| | - Nicole Monnier
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France.,Centre Hospitalier Régional Universitaire de Grenoble, Hôpital Michallon, Biochimie Génétique et Moléculaire, Grenoble, France
| | - Renée Krivosic
- Département Anesthésie-Réanimation, Hôpital Roger Salengro, CHRU de Lille, Lille, France
| | - Paul Allen
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, Davis CA, USA
| | - Arnaud Lacour
- Service de Neurologie, Hôpital Roger Salengro, CHRU de Lille, Lille, France
| | - Joël Lunardi
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France.,Centre Hospitalier Régional Universitaire de Grenoble, Hôpital Michallon, Biochimie Génétique et Moléculaire, Grenoble, France
| | - Julien Fauré
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France.,Centre Hospitalier Régional Universitaire de Grenoble, Hôpital Michallon, Biochimie Génétique et Moléculaire, Grenoble, France
| | - Isabelle Marty
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France.,Université Joseph Fourier, Grenoble, France
| |
Collapse
|
16
|
Intramolecular ex vivo Fluorescence Resonance Energy Transfer (FRET) of Dihydropyridine Receptor (DHPR) β1a Subunit Reveals Conformational Change Induced by RYR1 in Mouse Skeletal Myotubes. PLoS One 2015; 10:e0131399. [PMID: 26114725 PMCID: PMC4482598 DOI: 10.1371/journal.pone.0131399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 06/02/2015] [Indexed: 11/29/2022] Open
Abstract
The dihydropyridine receptor (DHPR) β1a subunit is essential for skeletal muscle excitation-contraction coupling, but the structural organization of β1a as part of the macromolecular DHPR-ryanodine receptor type I (RyR1) complex is still debatable. We used fluorescence resonance energy transfer (FRET) to probe proximity relationships within the β1a subunit in cultured skeletal myotubes lacking or expressing RyR1. The fluorescein biarsenical reagent FlAsH was used as the FRET acceptor, which exhibits fluorescence upon binding to specific tetracysteine motifs, and enhanced cyan fluorescent protein (CFP) was used as the FRET donor. Ten β1a reporter constructs were generated by inserting the CCPGCC FlAsH binding motif into five positions probing the five domains of β1a with either carboxyl or amino terminal fused CFP. FRET efficiency was largest when CCPGCC was positioned next to CFP, and significant intramolecular FRET was observed for all constructs suggesting that in situ the β1a subunit has a relatively compact conformation in which the carboxyl and amino termini are not extended. Comparison of the FRET efficiency in wild type to that in dyspedic (lacking RyR1) myotubes revealed that in only one construct (H458 CCPGCC β1a -CFP) FRET efficiency was specifically altered by the presence of RyR1. The present study reveals that the C-terminal of the β1a subunit changes conformation in the presence of RyR1 consistent with an interaction between the C-terminal of β1a and RyR1 in resting myotubes.
Collapse
|
17
|
Marty I. Triadin regulation of the ryanodine receptor complex. J Physiol 2014; 593:3261-6. [PMID: 26228554 DOI: 10.1113/jphysiol.2014.281147] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/19/2014] [Indexed: 11/08/2022] Open
Abstract
The calcium release complex is the major player in excitation-contraction coupling, both in cardiac and skeletal muscle. The core of the complex is the ryanodine receptor, and triadin is a regulating protein. Nevertheless, the precise function of triadin is only partially understood. Besides its function in the anchoring of calsequestrin at the triad/dyad, our recent results allow us to propose hypotheses on new triadin scaffolding functions, based on the studies performed using different models, from triadin knockout mice to human patients, and expression in non-muscle cells, taking into account the presence of multiple triadin isoforms.
Collapse
Affiliation(s)
- Isabelle Marty
- Grenoble Institut des Neurosciences, Inserm U836, Université Joseph Fourier-Bat EJ Safra, Chemin Fortuné Ferrini, 38700, La Tronche, France
| |
Collapse
|
18
|
Rendu J, Brocard J, Denarier E, Monnier N, Piétri-Rouxel F, Beley C, Roux-Buisson N, Gilbert-Dussardier B, Perez MJ, Romero N, Garcia L, Lunardi J, Fauré J, Fourest-Lieuvin A, Marty I. Exon skipping as a therapeutic strategy applied to an RYR1 mutation with pseudo-exon inclusion causing a severe core myopathy. Hum Gene Ther 2014; 24:702-13. [PMID: 23805838 DOI: 10.1089/hum.2013.052] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Central core disease is a myopathy often arising from mutations in the type 1 ryanodine receptor (RYR1) gene, encoding the sarcoplasmic reticulum calcium release channel RyR1. No treatment is currently available for this disease. We studied the pathological situation of a severely affected child with two recessive mutations, which resulted in a massive reduction in the amount of RyR1. The paternal mutation induced the inclusion of a new in-frame pseudo-exon in RyR1 mRNA that resulted in the insertion of additional amino acids leading to the instability of the protein. We hypothesized that skipping this additional exon would be sufficient to restore RyR1 expression and to normalize calcium releases. We therefore developed U7-AON lentiviral vectors to force exon skipping on affected primary muscle cells. The efficiency of the exon skipping was evaluated at the mRNA level, at the protein level, and at the functional level using calcium imaging. In these affected cells, we observed a decreased inclusion of the pseudo-exon, an increased RyR1 protein expression, and a restoration of calcium releases of normal amplitude either upon direct RyR1 stimulation or in response to membrane depolarization. This study is the first demonstration of the potential of exon-skipping strategy for the therapy of central core disease, from the molecular to the functional level.
Collapse
Affiliation(s)
- John Rendu
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, 38000 Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Congenital myopathies are severe muscle disorders affecting adults as well as children in all populations. The diagnosis of congenital myopathies is constrained by strong clinical and genetic heterogeneity. Moreover, the majority of patients present with unspecific histological features, precluding purposive molecular diagnosis and demonstrating the need for an alternative and more efficient diagnostic approach. We used exome sequencing complemented by histological and ultrastructural analysis of muscle biopsies to identify the causative mutations in eight patients with clinically different skeletal muscle pathologies, ranging from a fatal neonatal myopathy to a mild and slowly progressive myopathy with adult onset. We identified RYR1 (ryanodine receptor) mutations in six patients and NEB (nebulin) mutations in two patients. We found novel missense and nonsense mutations, unraveled small insertions/deletions and confirmed their impact on splicing and mRNA/protein stability. Histological and ultrastructural findings of the muscle biopsies of the patients validated the exome sequencing results. We provide the evidence that an integrated strategy combining exome sequencing with clinical and histopathological investigations overcomes the limitations of the individual approaches to allow a fast and efficient diagnosis, accelerating the patient's access to a better healthcare and disease management. This is of particular interest for the diagnosis of congenital myopathies, which involve very large genes like RYR1 and NEB as well as genetic and phenotypic heterogeneity.
Collapse
|
20
|
SERCA2a gene transfer prevents intimal proliferation in an organ culture of human internal mammary artery. Gene Ther 2012; 20:396-406. [PMID: 22763406 PMCID: PMC3465616 DOI: 10.1038/gt.2012.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Coronary restenosis, a major complication of percutaneous balloon angioplasty, results from neointimal proliferation of vascular smooth muscle cells (VSMCs). The sarco/endoplasmic reticulum calcium ATPase isoform 2a (SERCA2a), specific to contractile VSMCs, has been reported previously to be involved in the control of the Ca2+-signaling pathways governing proliferation and migration. Moreover, SERCA2a gene transfer was reported to inhibit in vitro VSMC proliferation and to prevent neointimal thickening in a rat carotid injury model. The aim of this study was to evaluate the potential therapeutic interest of SERCA2a gene transfer for prevention of in-stent restenosis using a human ex vivo model of left internal mammary artery (hIMA) intimal thickening. Left hIMAs, obtained at the time of aorto-coronary bypass surgeries, were subjected to balloon dilatation followed by infection for 30 min with adenoviruses encoding either human SERCA2 and GFP or control gene (beta-galactosidase) and GFP. Proliferation of subendothelial VSMCs and neointimal thickening were observed in balloon-injured hIMA maintained 14 days in organ culture under constant pressure and perfusion. SERCA2a gene transfer prevented vascular remodeling and significantly (p<0.01, n=5) reduced neointimal thickening in injured arteries (intima/media ratio was 0.07 ± 0.01 vs 0.40 ± 0.03 in βGal-infected arteries). These findings could have potential implications for treatment of pathological in stent-restenosis.
Collapse
|
21
|
Fourest-Lieuvin A, Rendu J, Osseni A, Pernet-Gallay K, Rossi D, Oddoux S, Brocard J, Sorrentino V, Marty I, Fauré J. Role of triadin in the organization of reticulum membrane at the muscle triad. J Cell Sci 2012; 125:3443-53. [PMID: 22505613 DOI: 10.1242/jcs.100958] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The terminal cisternae represent one of the functional domains of the skeletal muscle sarcoplasmic reticulum (SR). They are closely apposed to plasma membrane invaginations, the T-tubules, with which they form structures called triads. In triads, the physical interaction between the T-tubule-anchored voltage-sensing channel DHPR and the SR calcium channel RyR1 is essential because it allows the depolarization-induced calcium release that triggers muscle contraction. This interaction between DHPR and RyR1 is based on the peculiar membrane structures of both T-tubules and SR terminal cisternae. However, little is known about the molecular mechanisms governing the formation of SR terminal cisternae. We have previously shown that ablation of triadins, a family of SR transmembrane proteins that interact with RyR1, induced skeletal muscle weakness in knockout mice as well as a modification of the shape of triads. Here we explore the intrinsic molecular properties of the longest triadin isoform Trisk 95. We show that when ectopically expressed, Trisk 95 can modulate reticulum membrane morphology. The membrane deformations induced by Trisk 95 are accompanied by modifications of the microtubule network organization. We show that multimerization of Trisk 95 by disulfide bridges, together with interaction with microtubules, are responsible for the ability of Trisk 95 to structure reticulum membrane. When domains responsible for these molecular properties are deleted, anchoring of Trisk 95 to the triads in muscle cells is strongly decreased, suggesting that oligomers of Trisk 95 and microtubules contribute to the organization of the SR terminal cisternae in a triad.
Collapse
Affiliation(s)
- Anne Fourest-Lieuvin
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble 38042, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Roux-Buisson N, Cacheux M, Fourest-Lieuvin A, Fauconnier J, Brocard J, Denjoy I, Durand P, Guicheney P, Kyndt F, Leenhardt A, Le Marec H, Lucet V, Mabo P, Probst V, Monnier N, Ray PF, Santoni E, Trémeaux P, Lacampagne A, Fauré J, Lunardi J, Marty I. Absence of triadin, a protein of the calcium release complex, is responsible for cardiac arrhythmia with sudden death in human. Hum Mol Genet 2012; 21:2759-67. [PMID: 22422768 PMCID: PMC3363337 DOI: 10.1093/hmg/dds104] [Citation(s) in RCA: 190] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited arrhythmogenic disease so far related to mutations in the cardiac ryanodine receptor (RYR2) or the cardiac calsequestrin (CASQ2) genes. Because mutations in RYR2 or in CASQ2 are not retrieved in all CPVT cases, we searched for mutations in the physiological protein partners of RyR2 and CSQ2 in a large cohort of CPVT patients with no detected mutation in these two genes. Based on a candidate gene approach, we focused our investigations on triadin and junctin, two proteins that link RyR2 and CSQ2. Mutations in the triadin (TRDN) and in the junctin (ASPH) genes were searched in a cohort of 97 CPVT patients. We identified three mutations in triadin which cosegregated with the disease on a recessive mode of transmission in two families, but no mutation was found in junctin. Two TRDN mutations, a 4 bp deletion and a nonsense mutation, resulted in premature stop codons; the third mutation, a p.T59R missense mutation, was further studied. Expression of the p.T59R mutant in COS-7 cells resulted in intracellular retention and degradation of the mutant protein. This was confirmed after in vivo expression of the mutant triadin in triadin knock-out mice by viral transduction. In this work, we identified TRDN as a new gene responsible for an autosomal recessive form of CPVT. The mutations identified in the two families lead to the absence of the protein, thereby demonstrating the importance of triadin for the normal function of the cardiac calcium release complex in humans.
Collapse
Affiliation(s)
- Nathalie Roux-Buisson
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Golini L, Chouabe C, Berthier C, Cusimano V, Fornaro M, Bonvallet R, Formoso L, Giacomello E, Jacquemond V, Sorrentino V. Junctophilin 1 and 2 proteins interact with the L-type Ca2+ channel dihydropyridine receptors (DHPRs) in skeletal muscle. J Biol Chem 2011; 286:43717-43725. [PMID: 22020936 DOI: 10.1074/jbc.m111.292755] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Junctophilins (JPs) anchor the endo/sarcoplasmic reticulum to the plasma membrane, thus contributing to the assembly of junctional membrane complexes in striated muscles and neurons. Recent studies have shown that JPs may be also involved in regulating Ca2+ homeostasis. Here, we report that in skeletal muscle, JP1 and JP2 are part of a complex that, in addition to ryanodine receptor 1 (RyR1), includes caveolin 3 and the dihydropyridine receptor (DHPR). The interaction between JPs and DHPR was mediated by a region encompassing amino acids 230-369 and amino acids 216-399 in JP1 and JP2, respectively. Immunofluorescence studies revealed that the pattern of DHPR and RyR signals in C2C12 cells knocked down for JP1 and JP2 was rather diffused and characterized by smaller puncta in contrast to that observed in control cells. Functional experiments revealed that down-regulation of JPs in differentiated C2C12 cells resulted in a reduction of intramembrane charge movement and the L-type Ca2+ current accompanied by a reduced number of DHPRs at the plasma membrane, whereas there was no substantial alteration in Ca2+ release from the sterol regulatory element-binding protein. Altogether, these results suggest that JP1 and JP2 can facilitate the assembly of DHPR with other proteins of the excitation-contraction coupling machinery.
Collapse
Affiliation(s)
- Lucia Golini
- Molecular Medicine Section, Department of Neuroscience, and Interuniversity Institute of Myology, University of Siena, 53100 Siena, Italy
| | - Christophe Chouabe
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon 1, UMR CNRS 5534, 69622 Villeurbanne cedex, France
| | - Christine Berthier
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon 1, UMR CNRS 5534, 69622 Villeurbanne cedex, France
| | - Vincenza Cusimano
- Molecular Medicine Section, Department of Neuroscience, and Interuniversity Institute of Myology, University of Siena, 53100 Siena, Italy
| | - Mara Fornaro
- Molecular Medicine Section, Department of Neuroscience, and Interuniversity Institute of Myology, University of Siena, 53100 Siena, Italy
| | - Robert Bonvallet
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon 1, UMR CNRS 5534, 69622 Villeurbanne cedex, France
| | - Luca Formoso
- Molecular Medicine Section, Department of Neuroscience, and Interuniversity Institute of Myology, University of Siena, 53100 Siena, Italy
| | - Emiliana Giacomello
- Molecular Medicine Section, Department of Neuroscience, and Interuniversity Institute of Myology, University of Siena, 53100 Siena, Italy
| | - Vincent Jacquemond
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Lyon 1, UMR CNRS 5534, 69622 Villeurbanne cedex, France
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Neuroscience, and Interuniversity Institute of Myology, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
24
|
Trisk 32 regulates IP(3) receptors in rat skeletal myoblasts. Pflugers Arch 2011; 462:599-610. [PMID: 21811790 DOI: 10.1007/s00424-011-1001-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 01/25/2023]
Abstract
To date, four isoforms of triadins have been identified in rat skeletal muscle. While the function of the 95-kDa isoform in excitation-contraction coupling has been studied in detail, the role of the 32-kDa isoform (Trisk 32) remains elusive. Here, Trisk 32 overexpression was carried out by stable transfection in L6.G8 myoblasts. Co-localization of Trisk 32 and IP(3) receptors (IP(3)R) was demonstrated by immunocytochemistry, and their association was shown by co-immunoprecipitation. Functional effects of Trisk 32 on IP(3)-mediated Ca(2+) release were assessed by measuring changes in [Ca(2+)](i) following the stimulation by bradykinin or vasopressin. The amplitude of the Ca(2+) transients evoked by 20 μM bradykinin was significantly higher in Trisk 32-overexpressing (p < 0.01; 426 ± 84 nM, n = 27) as compared to control cells (76 ± 12 nM, n = 23). The difference remained significant (p < 0.02; 217 ± 41 nM, n = 21, and 97 ± 29 nM, n = 31, respectively) in the absence of extracellular Ca(2+). Similar observations were made when 0.1 μM vasopressin was used to initiate Ca(2+) release. Possible involvement of the ryanodine receptors (RyR) in these processes was excluded, after functional and biochemical experiments. Furthermore, Trisk 32 overexpression had no effect on store-operated Ca(2+) entry, despite a decrease in the expression of STIM1. These results suggest that neither the increased activity of RyR, nor the amplification of SOCE, is responsible for the differences observed in bradykinin- or vasopressin-evoked Ca(2+) transients; rather, they were due to the enhanced activity of IP(3)R. Thus, Trisk 32 not only co-localizes with, but directly contributes to, the regulation of Ca(2+) release via IP(3)R.
Collapse
|
25
|
Bevilacqua JA, Monnier N, Bitoun M, Eymard B, Ferreiro A, Monges S, Lubieniecki F, Taratuto AL, Laquerrière A, Claeys KG, Marty I, Fardeau M, Guicheney P, Lunardi J, Romero NB. Recessive RYR1 mutations cause unusual congenital myopathy with prominent nuclear internalization and large areas of myofibrillar disorganization. Neuropathol Appl Neurobiol 2011; 37:271-84. [PMID: 21062345 DOI: 10.1111/j.1365-2990.2010.01149.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS To report the clinical, pathological and genetic findings in a group of patients with a previously not described phenotype of congenital myopathy due to recessive mutations in the gene encoding the type 1 muscle ryanodine receptor channel (RYR1). METHODS Seven unrelated patients shared a predominant axial and proximal weakness of varying severity, with onset during the neonatal period, associated with bilateral ptosis and ophthalmoparesis, and unusual muscle biopsy features at light and electron microscopic levels. RESULTS Muscle biopsy histochemistry revealed a peculiar morphological pattern characterized by numerous internalized myonuclei in up to 51% of fibres and large areas of myofibrillar disorganization with undefined borders. Ultrastructurally, such areas frequently occupied the whole myofibre cross section and extended to a moderate number of sarcomeres in length. Molecular genetic investigations identified recessive mutations in the ryanodine receptor (RYR1) gene in six compound heterozygous patients and one homozygous patient. Nine mutations are novel and four have already been reported either as pathogenic recessive mutations or as changes affecting a residue associated with dominant malignant hyperthermia susceptibility. Only two mutations were located in the C-terminal transmembrane domain whereas the others were distributed throughout the cytoplasmic region of RyR1. CONCLUSION Our data enlarge the spectrum of RYR1 mutations and highlight their clinical and morphological heterogeneity. A congenital myopathy featuring ptosis and external ophthalmoplegia, concomitant with the novel histopathological phenotype showing fibres with large, poorly delimited areas of myofibrillar disorganization and internal nuclei, is highly suggestive of an RYR1-related congenital myopathy.
Collapse
Affiliation(s)
- J A Bevilacqua
- Institut de Myologie, Unité de Morphologie Neuromusculaire, Groupe Hospitalier-Universitaire Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bobe R, Hadri L, Lopez JJ, Sassi Y, Atassi F, Karakikes I, Liang L, Limon I, Lompré AM, Hatem SN, Hajjar RJ, Lipskaia L. SERCA2a controls the mode of agonist-induced intracellular Ca2+ signal, transcription factor NFAT and proliferation in human vascular smooth muscle cells. J Mol Cell Cardiol 2011; 50:621-33. [PMID: 21195084 PMCID: PMC3062203 DOI: 10.1016/j.yjmcc.2010.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 11/23/2010] [Accepted: 12/21/2010] [Indexed: 01/05/2023]
Abstract
In blood vessels, tone is maintained by agonist-induced cytosolic Ca(2+) oscillations of quiescent/contractile vascular smooth muscle cells (VSMCs). However, in synthetic/proliferative VSMCs, Gq/phosphoinositide receptor-coupled agonists trigger a steady-state increase in cytosolic Ca(2+) followed by a Store Operated Calcium Entry (SOCE) which translates into activation of the proliferation-associated transcription factor NFAT. Here, we report that in human coronary artery smooth muscle cells (hCASMCs), the sarco/endoplasmic reticulum calcium ATPase type 2a (SERCA2a) expressed in the contractile form of the hCASMCs, controls the nature of the agonist-induced Ca(2+) transient and the resulting down-stream signaling pathway. Indeed, restoring SERCA2a expression by gene transfer in synthetic hCASMCs 1) increased Ca(2+) storage capacity; 2) modified agonist-induced IP(3)R Ca(2+) release from steady-state to oscillatory mode (the frequency of agonist-induced IP(3)R Ca(2+) signal was 11.66 ± 1.40/100 s in SERCA2a-expressing cells (n=39) vs 1.37 ± 0.20/100 s in control cells (n=45), p<0.01); 3) suppressed SOCE by preventing interactions between SR calcium sensor STIM1 and pore forming unit ORAI1; 4) inhibited calcium regulated transcription factor NFAT and its down-stream physiological function such as proliferation and migration. This study provides evidence for the first time that oscillatory and steady-state patterns of Ca(2+) transients have different effects on calcium-dependent physiological functions in smooth muscle cells.
Collapse
Affiliation(s)
- Regis Bobe
- INSERM U770; Univ Paris Sud, Le Kremlin-Bicêtre, 94276, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Vassilopoulos S, Oddoux S, Groh S, Cacheux M, Fauré J, Brocard J, Campbell KP, Marty I. Caveolin 3 is associated with the calcium release complex and is modified via in vivo triadin modification. Biochemistry 2010; 49:6130-5. [PMID: 20565104 PMCID: PMC2907096 DOI: 10.1021/bi100796v] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The triadin isoforms Trisk 95 and Trisk 51 are both components of the skeletal muscle calcium release complex. To investigate the specific role of Trisk 95 and Trisk 51 isoforms in muscle physiology, we overexpressed Trisk 95 or Trisk 51 using adenovirus-mediated gene transfer in skeletal muscle of newborn mice. Overexpression of either Trisk 95 or Trisk 51 alters the muscle fiber morphology, while leaving unchanged the expression of the ryanodine receptor, the dihydropyridine receptor, and calsequestrin. We also observe an aberrant expression of caveolin 3 in both Trisk 95- and Trisk 51-overexpressing skeletal muscles. Using a biochemical approach, we demonstrate that caveolin 3 is associated with the calcium release complex in skeletal muscle. Taking advantage of muscle and non-muscle cell culture models and triadin null mouse skeletal muscle, we further dissect the molecular organization of the caveolin 3-containing calcium release complex. Our data demonstrate that the association of caveolin 3 with the calcium release complex occurs via a direct interaction with the transmembrane domain of the ryanodine receptor. Taken together, these data suggest that caveolin 3-containing membrane domains and the calcium release complex are functionally linked and that Trisk 95 and Trisk 51 are instrumental to the regulation of this interaction, the integrity of which may be crucial for muscle physiology.
Collapse
Affiliation(s)
- Stéphane Vassilopoulos
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Alteration of sarcoplasmic reticulum ca release in skeletal muscle from calpain 3-deficient mice. Int J Cell Biol 2010; 2009:340346. [PMID: 20300593 PMCID: PMC2838219 DOI: 10.1155/2009/340346] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 12/02/2009] [Indexed: 12/02/2022] Open
Abstract
Mutations of Ca2+-activated proteases (calpains) cause muscular dystrophies. Nevertheless, the specific role of calpains in Ca2+ signalling during the onset of dystrophies remains unclear. We investigated Ca2+ handling in skeletal cells from calpain 3-deficient mice. [Ca2+]i responses to caffeine, a ryanodine receptor (RyR) agonist, were decreased in −/− myotubes and absent in −/− myoblasts. The −/− myotubes displayed smaller amplitudes of the Ca2+ transients induced by cyclopiazonic acid in comparison to wild type cells. Inhibition of L-type Ca2+ channels (LCC) suppressed the caffeine-induced [Ca2+]i responses in −/− myotubes. Hence, the absence of calpain 3 modifies the sarcoplasmic reticulum (SR) Ca2+ release, by a decrease of the SR content, an impairment of RyR signalling, and an increase of LCC activity. We propose that calpain 3-dependent proteolysis plays a role in activating support proteins of intracellular Ca2+ signalling at a stage of cellular differentiation which is crucial for skeletal muscle regeneration.
Collapse
|
29
|
Oddoux S, Brocard J, Schweitzer A, Szentesi P, Giannesini B, Brocard J, Fauré J, Pernet-Gallay K, Bendahan D, Lunardi J, Csernoch L, Marty I. Triadin deletion induces impaired skeletal muscle function. J Biol Chem 2009; 284:34918-29. [PMID: 19843516 PMCID: PMC2787354 DOI: 10.1074/jbc.m109.022442] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 10/14/2009] [Indexed: 01/27/2023] Open
Abstract
Triadin is a multiple proteins family, some isoforms being involved in muscle excitation-contraction coupling, and some having still unknown functions. To obtain clues on triadin functions, we engineered a triadin knock-out mouse line and characterized the physiological effect of triadin ablation on skeletal muscle function. These mice presented a reduced muscle strength, which seemed not to alter their survival and has been characterized in the present work. We first checked in these mice the expression level of the different proteins involved in calcium homeostasis and observed in fast muscles an increase in expression of dihydropyridine receptor, with a large reduction in calsequestrin expression. Electron microscopy analysis of KO muscles morphology demonstrated the presence of triads in abnormal orientation and a reduction in the sarcoplasmic reticulum terminal cisternae volume. Using calcium imaging on cultured myotubes, we observed a reduction in the total amount of calcium stored in the sarcoplasmic reticulum. Physiological studies have been performed to evaluate the influence of triadin deletion on skeletal muscle function. Muscle strength has been measured both on the whole animal model, using hang test or electrical stimulation combined with NMR analysis and strength measurement, or on isolated muscle using electrical stimulation. All the results obtained demonstrate an important reduction in muscle strength, indicating that triadin plays an essential role in skeletal muscle function and in skeletal muscle structure. These results indicate that triadin alteration leads to the development of a myopathy, which could be studied using this new animal model.
Collapse
Affiliation(s)
- Sarah Oddoux
- From INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble F-38000, France
- the Université Joseph Fourier, Grenoble F-38000, France
| | - Julie Brocard
- From INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble F-38000, France
- the Université Joseph Fourier, Grenoble F-38000, France
| | - Annie Schweitzer
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Physiopathologie du Cytosquelette, Grenoble F-38000, France
| | - Peter Szentesi
- the Department of Physiology, Medical School and Health Science Center, University of Debrecen, H-4012 Debrecen, Hungary
| | - Benoit Giannesini
- the Centre de Résonance Magnétique Biologique et Médicale, UMR CNRS 6612, Faculté de Médecine de la Timone, Marseille 13000, France, and
| | - Jacques Brocard
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Physiopathologie du Cytosquelette, Grenoble F-38000, France
| | - Julien Fauré
- From INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble F-38000, France
- the Université Joseph Fourier, Grenoble F-38000, France
- Centre Hospitalier Regional Universitaire de Grenoble, Hopital Michallon, Biochimie et Génétique Moléculaire, Grenoble F-38000, France
| | - Karine Pernet-Gallay
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Physiopathologie du Cytosquelette, Grenoble F-38000, France
| | - David Bendahan
- the Centre de Résonance Magnétique Biologique et Médicale, UMR CNRS 6612, Faculté de Médecine de la Timone, Marseille 13000, France, and
| | - Joël Lunardi
- From INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble F-38000, France
- the Université Joseph Fourier, Grenoble F-38000, France
- Centre Hospitalier Regional Universitaire de Grenoble, Hopital Michallon, Biochimie et Génétique Moléculaire, Grenoble F-38000, France
| | - Laszlo Csernoch
- the Department of Physiology, Medical School and Health Science Center, University of Debrecen, H-4012 Debrecen, Hungary
| | - Isabelle Marty
- From INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble F-38000, France
- the Université Joseph Fourier, Grenoble F-38000, France
| |
Collapse
|
30
|
Monnier N, Laquerrière A, Marret S, Goldenberg A, Marty I, Nivoche Y, Lunardi J. First genomic rearrangement of the RYR1 gene associated with an atypical presentation of lethal neonatal hypotonia. Neuromuscul Disord 2009; 19:680-4. [PMID: 19734047 DOI: 10.1016/j.nmd.2009.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 07/08/2009] [Accepted: 07/16/2009] [Indexed: 11/30/2022]
Abstract
Neonatal hypotonia is frequently observed with a highly variable clinical presentation. Congenital myopathies that are classically characterized by the presence of structural changes of the muscle fibres such as cores, rods and aggregates have been reported to be occasionally associated with this presentation. However, the identification of the causing defect can be a challenging task in severe neonatal forms of the disease since specific structural changes might not always be present in affected newborn's muscles. The RYR1 gene encodes the skeletal muscle isoform of a calcium channel, the ryanodine receptor, and has been involved in both dominant and recessive congenital myopathies associated with structural changes and presenting with various degree of severity. Here we report the case of a child presenting at birth with a lethal form of neonatal hypotonia associated with an atypical congenital myopathy. Molecular investigations showed that the disease was caused by two novel RYR1 mutations. One of the mutations was a large-sized genomic deletion. This is the first genomic rearrangement identified into the RYR1 gene to our knowledge. This new class of mutation of the RYR1 gene will clearly have consequences for the molecular investigation of RYR1-related diseases.
Collapse
Affiliation(s)
- Nicole Monnier
- Laboratoire de Biochimie et Génétique Moléculaire, CHU de Grenoble, Grenoble, France
| | | | | | | | | | | | | |
Collapse
|
31
|
Marty I, Fauré J, Fourest-Lieuvin A, Vassilopoulos S, Oddoux S, Brocard J. Triadin: what possible function 20 years later? J Physiol 2009; 587:3117-21. [PMID: 19403623 DOI: 10.1113/jphysiol.2009.171892] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
During the last 20 years, the identification of triadin function in cardiac and skeletal muscle has been the focus of numerous studies. First thought of as the missing link between the ryanodine receptor and the dihydropyridine receptor and responsible of skeletal type excitation-contraction coupling, the current hypothesis on triadin function has slowly evolved, and triadin is envisaged now as a regulator of calcium release, both in cardiac and skeletal muscle. Nevertheless, none of the experiments performed up to now has given a clear cut view of what triadin really does in muscle. The problem became more complex with the identification of multiple triadin isoforms, having possibly multiple functions. Using a different approach from what has been done previously, we have obtained new clues about the function of triadin. Our data point to a possible involvement of triadin in reticulum structure, in relation with the microtubule network.
Collapse
|
32
|
Rigoard P, Buffenoir K, Wager M, Bauche S, Giot JP, Lapierre F. [Molecular architecture of the sarcoplasmic reticulum and its role in the ECC]. Neurochirurgie 2009; 55 Suppl 1:S83-91. [PMID: 19233437 DOI: 10.1016/j.neuchi.2008.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Accepted: 05/09/2008] [Indexed: 11/16/2022]
Abstract
The sarcoplasmic reticulum (SR) plays a fundamental role in excitation-contraction coupling, which propagates the electric signal conversion along the muscle fiber's plasmic membrane to a mechanical event manifested as a muscle contraction. It plays a crucial role in calcium homeostasis and intracellular calcium storage control (storage, liberation and uptake) necessary for fiber muscle contraction and then relaxation. These functions take place at the triad, made up of individualized SR subdomains where the protein-specific organization provides efficient and fast coupling. Ryanodine receptors (RyR) and dihydropyridine receptors (DHPR) mainly act in calcium exchanges in the SR. This particular structural and molecular architecture must be correlated to its functional specificity.
Collapse
Affiliation(s)
- P Rigoard
- Service de neurochirurgie, CHU La Milétrie, 2, rue de la Milétrie, BP 577, 86021 Poitiers cedex, France.
| | | | | | | | | | | |
Collapse
|
33
|
Shen X, Franzini-Armstrong C, Lopez JR, Jones LR, Kobayashi YM, Wang Y, Kerrick WGL, Caswell AH, Potter JD, Miller T, Allen PD, Perez CF. Triadins modulate intracellular Ca(2+) homeostasis but are not essential for excitation-contraction coupling in skeletal muscle. J Biol Chem 2007; 282:37864-74. [PMID: 17981799 DOI: 10.1074/jbc.m705702200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To unmask the role of triadin in skeletal muscle we engineered pan-triadin-null mice by removing the first exon of the triadin gene. This resulted in a total lack of triadin expression in both skeletal and cardiac muscle. Triadin knockout was not embryonic or birth-lethal, and null mice presented no obvious functional phenotype. Western blot analysis of sarcoplasmic reticulum (SR) proteins in skeletal muscle showed that the absence of triadin expression was associated with down-regulation of Junctophilin-1, junctin, and calsequestrin but resulted in no obvious contractile dysfunction. Ca(2+) imaging studies in null lumbricalis muscles and myotubes showed that the lack of triadin did not prevent skeletal excitation-contraction coupling but reduced the amplitude of their Ca(2+) transients. Additionally, null myotubes and adult fibers had significantly increased myoplasmic resting free Ca(2+).[(3)H]Ryanodine binding studies of skeletal muscle SR vesicles detected no differences in Ca(2+) activation or Ca(2+) and Mg(2+) inhibition between wild-type and triadin-null animals. Subtle ultrastructural changes, evidenced by the appearance of longitudinally oriented triads and the presence of calsequestrin in the sacs of the longitudinal SR, were present in fast but not slow twitch-null muscles. Overall, our data support an indirect role for triadin in regulating myoplasmic Ca(2+) homeostasis and organizing the molecular complex of the triad but not in regulating skeletal-type excitation-contraction coupling.
Collapse
Affiliation(s)
- Xiaohua Shen
- Department of Anesthesiology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Divet A, Paesante S, Grasso C, Cavagna D, Tiveron C, Paolini C, Protasi F, Huchet-Cadiou C, Treves S, Zorzato F. Increased Ca2+ storage capacity of the skeletal muscle sarcoplasmic reticulum of transgenic mice over-expressing membrane bound calcium binding protein junctate. J Cell Physiol 2007; 213:464-74. [PMID: 17516551 DOI: 10.1002/jcp.21121] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Junctate is an integral sarco(endo)plasmic reticulum protein expressed in many tissues including heart and skeletal muscle. Because of its localization and biochemical characteristics, junctate is deemed to participate in the regulation of the intracellular Ca2+ concentration. However, its physiological function in muscle cells has not been investigated yet. In this study we examined the effects of junctate over-expression by generating a transgenic mouse model which over-expresses junctate in skeletal muscle. Our results demonstrate that junctate over-expression induced a significant increase in SR Ca2+ storage capacity which was paralleled by an increased 4-chloro-m-cresol and caffeine-induced Ca2+ release, whereas it did not affect SR Ca2+-dependent ATPase activity and SR Ca2+ loading rates. In addition, junctate over-expression did not affect the expression levels of SR Ca2+ binding proteins such as calsequestrin, calreticulin and sarcalumenin. These findings suggest that junctate over-expression is associated with an increase in the SR Ca2+ storage capacity and releasable Ca2+ content and support a physiological role for junctate in intracellular Ca2+ homeostasis.
Collapse
Affiliation(s)
- Alexandra Divet
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yang T, Allen PD, Pessah IN, Lopez JR. Enhanced excitation-coupled calcium entry in myotubes is associated with expression of RyR1 malignant hyperthermia mutations. J Biol Chem 2007; 282:37471-8. [PMID: 17942409 DOI: 10.1074/jbc.m701379200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myotubes expressing wild type RyR1 (WT) or RyR1 with one of three malignant hyperthermia mutations R615C, R2163C, and T4826I (MH) were exposed sequentially to 60 mm KCl in Ca(2+)-replete and Ca(2+)-free external buffers (Ca+ and Ca-, respectively) with 3 min of rest between exposures. Although the maximal peak amplitude of the Ca(2+) transients during K(+) depolarization was similar for WT and MH in both external buffers, the rate of decay of the sustained phase of the transient during K(+) depolarization (decay rate) in Ca+ was 50% slower for MH. This difference was eliminated in Ca-, and the relative decay rates were faster for both genotypes than in Ca+. The integrated Ca(2+) transient in Ca-compared with Ca+ was reduced by 50-60% for MH and 20% for WT. The decay rate was not affected by [K(+)] x [Cl(-)] product or NiCl(2) (2 mm) supplementation of Ca-. The addition of La(2+) (0.1 mm), or SKF 96365 (20 microm) to Ca+ significantly accelerated decay rates for both WT and MH, but their effect was significantly greater in MH. Nifedipine (1 microm) had no effect, suggesting that the mechanism for this difference was not a reduction in L-type Ca(2+) channel Ca(2+) current. These data strongly suggest: 1) the decay rate in skeletal myotubes is related in part to Ca(2+) entry through the ECCE channel; 2) the MH mutations enhance ECCE compared with wild type; and 3) the increased Ca(2+) entry might play a significant role in the pathophysiology of MH.
Collapse
Affiliation(s)
- Tianzhong Yang
- Department of Anesthesiology Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
36
|
Kim S, Yun HM, Baik JH, Chung KC, Nah SY, Rhim H. Functional interaction of neuronal Cav1.3 L-type calcium channel with ryanodine receptor type 2 in the rat hippocampus. J Biol Chem 2007; 282:32877-89. [PMID: 17823125 DOI: 10.1074/jbc.m701418200] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal L-type Ca(2+) channels do not support synaptic transmission, but they play an essential role in synaptic activity-dependent gene expression. Ca(v)1.2 and Ca(v)1.3 are the two most widely expressed L-type Ca(2+) channels in neurons and have different biophysical and subcellular distributions. The function of the Ca(v) 1.3 L-type Ca(2+) channel and its cellular mechanisms in the central nervous system are poorly understood. In this study, using a yeast two-hybrid assay, we found that the N terminus of the rat Ca(v)1.3 alpha(1) subunit interacts with a partial N-terminal amino acid sequence of ryanodine receptor type 2 (RyR2). Reverse transcription-PCR and Western blot assays revealed high expression of both Ca(v)1.3 and RyR2 in the rat hippocampus. We also demonstrate a physical association of Ca(v)1.3 with RyR2 using co-immunoprecipitation assays. Moreover, immunocytochemistry revealed prominent co-localization between Ca(v)1.3 and RyR2 in hippocampal neurons. Depolarizing cells by an acute treatment of a high concentration of KCl (high-K, 60 mm) showed that the activation of L-type Ca(2+) channels induced RyR opening and led to RyR-dependent Ca(2+) release, even in the absence of extracellular Ca(2+). Furthermore, we found that RyR2 mRNA itself is increased by long term treatment of high-K via activation of L-type Ca(2+) channels. These acute and long term effects of high-K on RyRs were selectively blocked by small interfering RNA-mediated silencing of Ca(v)1.3. These results suggest a physical and functional interaction between Ca(v)1.3 and RyR2 and important implications of Ca(v)1.3/RyR2 clusters in translating synaptic activity into alterations in gene expression.
Collapse
Affiliation(s)
- Sunoh Kim
- Life Sciences Division, Korea Institute of Science and Technology, 39-1 Hawholgok-dong, Sungbuk-gu, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
37
|
Altafaj X, France J, Almassy J, Jona I, Rossi D, Sorrentino V, Mabrouk K, De Waard M, Ronjat M. Maurocalcine interacts with the cardiac ryanodine receptor without inducing channel modification. Biochem J 2007; 406:309-15. [PMID: 17537000 PMCID: PMC1948973 DOI: 10.1042/bj20070453] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have previously shown that MCa (maurocalcine), a toxin from the venom of the scorpion Maurus palmatus, binds to RyR1 (type 1 ryanodine receptor) and induces strong modifications of its gating behaviour. In the present study, we investigated the ability of MCa to bind to and modify the gating process of cardiac RyR2. By performing pull-down experiments we show that MCa interacts directly with RyR2 with an apparent affinity of 150 nM. By expressing different domains of RyR2 in vitro, we show that MCa binds to two domains of RyR2, which are homologous with those previously identified on RyR1. The effect of MCa binding to RyR2 was then evaluated by three different approaches: (i) [(3)H]ryanodine binding experiments, showing a very weak effect of MCa (up to 1 muM), (ii) Ca(2+) release measurements from cardiac sarcoplasmic reticulum vesicles, showing that MCa up to 1 muM is unable to induce Ca(2+) release, and (iii) single-channel recordings, showing that MCa has no effect on the open probability or on the RyR2 channel conductance level. Long-lasting opening events of RyR2 were observed in the presence of MCa only when the ionic current direction was opposite to the physiological direction, i.e. from the cytoplasmic face of RyR2 to its luminal face. Therefore, despite the conserved MCa binding ability of RyR1 and RyR2, functional studies show that, in contrast with what is observed with RyR1, MCa does not affect the gating properties of RyR2. These results highlight a different role of the MCa-binding domains in the gating process of RyR1 and RyR2.
Collapse
Affiliation(s)
- Xavier Altafaj
- *iRTSV/CCFP CEA Grenoble INSERM U836 Institut des Neurosciences Grenoble GIN, 17 rue des Martyrs, 38054 Grenoble Cedex 09, France
| | - Julien France
- *iRTSV/CCFP CEA Grenoble INSERM U836 Institut des Neurosciences Grenoble GIN, 17 rue des Martyrs, 38054 Grenoble Cedex 09, France
| | - Janos Almassy
- †Department of Physiology, Research Center of Molecular Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Istvan Jona
- †Department of Physiology, Research Center of Molecular Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Daniela Rossi
- ‡Molecular Medicine Section, Department of Neuroscience, University of Siena, Siena, Italy
| | - Vincenzo Sorrentino
- ‡Molecular Medicine Section, Department of Neuroscience, University of Siena, Siena, Italy
| | - Kamel Mabrouk
- §Universités D'Aix-Marseille 1, 2 et 3 CNRS-UMR 6517, Chimie, Biologie et Radicaux libres, Case 521Av.Esc. Normandie Niemen 13397 Marseille Cédex 20, France
| | - Michel De Waard
- *iRTSV/CCFP CEA Grenoble INSERM U836 Institut des Neurosciences Grenoble GIN, 17 rue des Martyrs, 38054 Grenoble Cedex 09, France
| | - Michel Ronjat
- *iRTSV/CCFP CEA Grenoble INSERM U836 Institut des Neurosciences Grenoble GIN, 17 rue des Martyrs, 38054 Grenoble Cedex 09, France
- To whom correspondence should be addressed (email )
| |
Collapse
|
38
|
Lipskaia L, Pinet C, Fromes Y, Hatem S, Cantaloube I, Coulombe A, Lompré AM. Mutation of delta-sarcoglycan is associated with Ca(2+) -dependent vascular remodeling in the Syrian hamster. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:162-71. [PMID: 17591963 PMCID: PMC1941595 DOI: 10.2353/ajpath.2007.070054] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We examined whether mutation of the delta-sarcoglycan gene, which causes dilated cardiomyopathy, also alters the vascular smooth muscle cell (VSMC) phenotype and arterial function in the Syrian hamster CHF 147. Thoracic aorta media thickness showed marked variability in diseased hamsters with zones of atrophy and hypertrophied segments. CHF-147 VSMCs displayed a proliferating/"synthetic" phenotype characterized by the absence of the smooth muscle myosin heavy chain SM2, dystrophin, and Ca(2+)-handling proteins, and the presence of cyclin D1. In freshly isolated VSMCs from CHF 147 hamsters, voltage-independent basal Ca(2+) channels showed enhanced activity similar to that in proliferating wild-type (WT) cells. The transcription factor NFAT (nuclear factor of activated T cells) was spontaneously active in freshly isolated CHF 147 VSMCs, as in proliferating VSMCs from WT hamsters. Mibefradil inhibited B-type channels, NFAT activity, and VSMC proliferation. CHF 147 hamsters had abundant apoptotic cells distributed in patches along the aorta, and clusters of inactive mitochondria were observed in 25% of isolated CHF 147 cells, whereas no such clusters were seen in WT cells. In conclusion, mutation of the delta-sarcoglycan gene increases plasma membrane permeability to Ca(2+), activates the Ca(2+)-regulated transcription factor NFAT, and leads to spontaneous mitochondrial aggregation, causing abnormal VSMC proliferation and apoptosis.
Collapse
Affiliation(s)
- Larissa Lipskaia
- INSERM UMR S621, 91 bd de l'Hôpital, 75634 Paris Cedex 13, France
| | | | | | | | | | | | | |
Collapse
|
39
|
Avila G, Aguilar CI, Ramos-Mondragón R. Sustained CGRP1 receptor stimulation modulates development of EC coupling by cAMP/PKA signalling pathway in mouse skeletal myotubes. J Physiol 2007; 584:47-57. [PMID: 17656431 PMCID: PMC2277057 DOI: 10.1113/jphysiol.2007.137687] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We investigated modulation of excitation-contraction (EC) coupling by calcitonin gene-related peptide (CGRP), which is released by motorneurons during neuromuscular transmission. Mouse skeletal myotubes were cultured either under control conditions or in the presence of 100 nm CGRP ( approximately 4-72 h). T- and L-type Ca(2+) currents, immobilization resistant charge movement, and intracellular Ca(2+) transients were characterized in whole-cell patch-clamp experiments. CGRP treatment increased the amplitude of voltage-gated Ca(2+) release ((DeltaF/F)(max)) approximately 75-350% and moderately increased both maximal L-current conductance (G(max)) and charge movement (Q(max)). In contrast, CGRP treatment did not affect their corresponding voltage dependence of activation (V(1/2) and k) or T-current density. CGRP treatment enhanced voltage-gated Ca(2+) release in approximately 4 h, whereas the effect on L-channel magnitude took longer to develop ( approximately 24 h), suggesting that short-term potentiation of EC coupling may lead to subsequent long-term up-regulation of DHPR expression. CGRP treatment also drastically increased caffeine-induced Ca(2+) release in approximately 4 h ( approximately 400%). Thus, short-term potentiation of EC coupling is due to an increase in sarcoplasmic reticulum Ca(2+) content. Both application of a phosphodiesterase inhibitor (papaverine) and a membrane-permeant cAMP analogue (Db-cAMP) produced a similar potentiation of EC coupling. Conversely, this potentiation was prevented by pretreatment with either CGRP1 receptor antagonist (CGRP(8-37)) or a PKA inhibitor (H-89). Thus, CGRP acts through CGRP1 receptors and the cAMP/PKA signalling pathway to enhance voltage-gated Ca(2+) release. Effects of CGRP on both EC coupling and L-channels were attenuated at later times during myotube differentiation. Therefore, we conclude that CGRP accelerates maturation of EC coupling.
Collapse
Affiliation(s)
- Guillermo Avila
- Departamento de Bioquímica, Cinvestav-IPN, AP 14-740, México, DF 07000, México.
| | | | | |
Collapse
|
40
|
Lyfenko AD, Ducreux S, Wang Y, Xu L, Zorzato F, Ferreiro A, Meissner G, Treves S, Dirksen RT. Two central core disease (CCD) deletions in the C-terminal region of RYR1 alter muscle excitation-contraction (EC) coupling by distinct mechanisms. Hum Mutat 2007; 28:61-8. [PMID: 16958053 DOI: 10.1002/humu.20409] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Central core disease (CCD) and malignant hyperthermia (MH) are skeletal muscle disorders that are linked to mutations in the gene that encodes the type 1 ryanodine receptor (RYR1). The RYR1 ion channel plays a central role in excitation-contraction (EC) coupling by releasing Ca(2+) from an internal store. Pathogenic CCD mutations in RYR1 result in changes in the magnitude of Ca(2+) release during EC coupling. CCD has recently been linked to two novel deletions (c.12640_12648delCGCCAGTTC [p.Arg4214_Phe4216del] and c.14779_14784delGTCATC [p.Val4927_Ile4928del]) in the C-terminal region of RYR1. To determine the phenotypic consequences of these mutations and extend our understanding of the pathogenic mechanisms that underlie CCD, we determined functional effects on Ca(2+) release channel activity of analogous deletions (p.Arg4215_Phe4217del and p.Val4926_Ile4927del) engineered into rabbit RYR1 following expression in RYR1-null (dyspedic) myotubes and HEK293 cells. In addition, we assessed effects of the p.Arg4214 Phe4216del mutation on RYR1 function in lymphoblastoid cells obtained from CCD patients heterozygous for the mutation. Here we report that both deletions significantly reduce Ca(2+) release following RYR1 activation, but by different mechanisms. While the p.Arg4214_Phe4216del deletion promotes Ca(2+) depletion from intracellular stores by exhibiting a classic "leaky channel" behavior, the p.Val4927_Ile4928del deletion reduces Ca(2+) release by disrupting Ca(2+) gating and eliminating Ca(2+) permeation through the open channel.
Collapse
Affiliation(s)
- Alla D Lyfenko
- Department of Physiology and Pharmacology, University of Rochester, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Han HM, Wei RS, Lai FA, Lai AF, Yin CC. Molecular nature of sulfhydryl modification by hydrogen peroxide on type 1 ryanodine receptor. Acta Pharmacol Sin 2006; 27:888-94. [PMID: 16787573 DOI: 10.1111/j.1745-7254.2006.00386.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM To elucidate the molecular nature of sulfhydryl modification by hydrogen peroxide on type 1 ryanodine receptor (RyR1). METHODS Rabbit skeletal muscle sarcoplasmic reticulum was treated with hydrogen peroxide, then RyR1 complex was isolated. The proteins in the complex were analysed by electrophoresis, Western blot and electron microscopy. RESULTS (1) Hydrogen peroxide induces inter-subunit cross-linking within the tetrameric RyR1 molecule; (2) in parallel to inter-subunit cross-linking, the RyR1 molecule changes morphology; (3) the chemical and morphological changes are reversible: upon reduction by reducing agents, the RyR1 molecule regains its original state. CONCLUSION These findings suggest that the molecular mechanism of RyR1 channel activity in sarcoplasmic reticulum regulated by hydrogen peroxide is through inter-subunit cross-linking within the tetrameric RyR1 molecule, which in turn induces structural changes of RyR1.
Collapse
Affiliation(s)
- Hong-mei Han
- Department of Biophysics, Peking University Health Science Centre, Peking University, Beijing 100083, China
| | | | | | | | | |
Collapse
|
42
|
Paydar MJ, Pousti A, Farsam H, Amanlou M, Mehr SE, Dehpour AR. Effects of diltiazem or verapamil on calcium uptake and release from chicken skeletal muscle sarcoplasmic reticulum. Can J Physiol Pharmacol 2006; 83:967-75. [PMID: 16391705 DOI: 10.1139/y05-062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to determine the effects of 2 Ca2+ channel blockers, verapamil and diltiazem, on calcium loading (active Ca2+ uptake) and the following Ca2+ release induced by silver ion (Ag+) and Ca2+ from the membrane of heavy sarcoplasmic reticulum (SR) of chicken skeletal muscle. A fluorescent probe technique was employed to determine the calcium movement through the SR. Pretreatment of the medium with diltiazem and verapamil resulted in a significant decrease in the active Ca2+ uptake, with IC50 of about 290 micromol/L for verapamil and 260 micromol/L for diltiazem. Inhibition of Ca2+ uptake was not due to the development of a substantial drug-dependent leak of Ca2+ from the SR. It might, in part, have been mediated by a direct inhibitory effect of these drugs on the Ca2+ ATPase activity of the SR Ca2+ pump. We confirmed that Ca2+ channel blockers, administered after SR Ca2+ loading and before induction of Ca2+ release, caused a dose-dependent inhibition of both Ca2+- and Ag+-induced Ca2+ release rate. Moreover, if Ca2+ channel blockers were administered prior to SR Ca2+ loading, in spite of Ca2+ uptake inhibition the same reduction in Ca2+- and Ag+-induced Ca2+ release rate was seen. We showed that the inhibition of Ag+-induced Ca2+ release by L-channel blockers is more sensitive than Ca2+-induced Ca2+ release inhibition, so the IC50 for Ag+- and Ca2+-induced Ca2+ release was about 100 and 310 micromol/L for verapamil and 79 and 330 micromol/L for diltiazem, respectively. Our results support the evidence that Ca2+ channel blockers affect muscle microsome of chicken skeletal muscle by 2 independent mechanisms: first, reduction of Ca2+ uptake rate and Ca2+-ATPase activity inhibition, and second, inhibition of both Ag+- and Ca2+-induced Ca2+ release by Ca2+ release channels. These findings confirm the direct effect of Ca2+ channel blockers on calcium release channels. Our results suggest that even if the SR is incompletely preloaded with Ca2+ because of inhibition of Ca2+ uptake by verapamil and diltiazem, no impairment in Ca2+ release occurs.
Collapse
Affiliation(s)
- Mehrak Javadi Paydar
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran
| | | | | | | | | | | |
Collapse
|
43
|
Divet A, Paesante S, Bleunven C, Anderson A, Treves S, Zorzato F. Novel sarco(endo)plasmic reticulum proteins and calcium homeostasis in striated muscles. J Muscle Res Cell Motil 2005; 26:7-12. [PMID: 16096683 DOI: 10.1007/s10974-005-9001-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Revised: 06/09/2005] [Accepted: 06/17/2005] [Indexed: 10/25/2022]
Abstract
The impact of calcium signaling on many cellular functions is reflected by the tight regulation of the intracellular Ca(2+) concentration that is ensured by diverse pumps, channels, transporters and Ca(2+) binding proteins. In this review, we present recently identified novel sarco(endo)plasmic reticulum proteins that may have a potential involvement in the regulation of Ca(2+) homeostasis in striated muscles.
Collapse
Affiliation(s)
- A Divet
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, University of Ferrara, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Magkos F, Kavouras SA. Caffeine Use in Sports, Pharmacokinetics in Man, and Cellular Mechanisms of Action. Crit Rev Food Sci Nutr 2005; 45:535-62. [PMID: 16371327 DOI: 10.1080/1040-830491379245] [Citation(s) in RCA: 221] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Caffeine is the most widely consumed psychoactive 'drug' in the world and probably one of the most commonly used stimulants in sports. This is not surprising, since it is one of the few ergogenic aids with documented efficiency and minimal side effects. Caffeine is rapidly and completely absorbed by the gastrointestinal tract and is readily distributed throughout all tissues of the body. Peak plasma concentrations after normal consumption are usually around 50 microM, and half-lives for elimination range between 2.5-10 h. The parent compound is extensively metabolized in the liver microsomes to more than 25 derivatives, while considerably less than 5% of the ingested dose is excreted unchanged in the urine. There is, however, considerable inter-individual variability in the handling of caffeine by the body, due to both environmental and genetic factors. Evidence from in vitro studies provides a wealth of different cellular actions that could potentially contribute to the observed effects of caffeine in humans in vivo. These include potentiation of muscle contractility via induction of sarcoplasmic reticulum calcium release, inhibition of phosphodiesterase isoenzymes and concomitant cyclic monophosphate accumulation, inhibition of glycogen phosphorylase enzymes in liver and muscle, non-selective adenosine receptor antagonism, stimulation of the cellular membrane sodium/potassium pump, impairment of phosphoinositide metabolism, as well as other, less thoroughly characterized actions. Not all, however, seem to account for the observed effects in vivo, although a variable degree of contribution cannot be readily discounted on the basis of experimental data. The most physiologically relevant mechanism of action is probably the blockade of adenosine receptors, but evidence suggests that, at least under certain conditions, other biochemical mechanisms may also be operational.
Collapse
Affiliation(s)
- Faidon Magkos
- Laboratory of Nutrition and Clinical Dietetics, Department of Nutrition and Dietetics, Harokopio University, 176 71 Kallithea, Athens, Greece
| | | |
Collapse
|
45
|
Rezgui SS, Vassilopoulos S, Brocard J, Platel JC, Bouron A, Arnoult C, Oddoux S, Garcia L, De Waard M, Marty I. Triadin (Trisk 95) overexpression blocks excitation-contraction coupling in rat skeletal myotubes. J Biol Chem 2005; 280:39302-8. [PMID: 16176928 PMCID: PMC2739420 DOI: 10.1074/jbc.m506566200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To identify the function of triadin in skeletal muscle, adenovirus-mediated overexpression of Trisk 95 or Trisk 51, the two major skeletal muscle isoforms, was induced in rat skeletal muscle primary cultures, and the physiological behavior of the modified cells was analyzed. Overexpression did not modify the expression level of their protein partners ryanodine receptor, dihydropyridine receptor, and the other triadin. Caffeine-induced calcium release was also unaffected by triadin overexpression. Nevertheless, in the absence of extracellular calcium, depolarization-induced calcium release was almost abolished in Trisk 95 overexpressing myotubes (T95 myotubes), and not modified in Trisk 51 overexpressing myotubes (T51 myotubes). This was not because of a modification of dihydropyridine receptors, as depolarization in presence of external calcium still induced a calcium release, and the activation curve of dihydropyridine receptor was unchanged, in both T95 and T51 myotubes. The calcium release complex was also maintained in T95 myotubes as Trisk 95, ryanodine receptor, dihydropyridine receptor, and Trisk 51 were still co-localized. The effect of Trisk 95 overexpression on depolarization-induced calcium release was reversed by a simultaneous infection with an antisense Trisk 95 adenovirus, indicating the specificity of this effect. Thus, the level of Trisk 95 and not Trisk 51 is important on regulating the calcium release complex, and an excess of this protein can lead to an inhibition of the physiological function of the complex.
Collapse
Affiliation(s)
- Sophia Smida Rezgui
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Stéphane Vassilopoulos
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Julie Brocard
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Jean Claude Platel
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Alexandre Bouron
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Christophe Arnoult
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Sarah Oddoux
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Luis Garcia
- Genethon
Genethon1 rue de l'Internationale, 91002 Evry,FR
| | - Michel De Waard
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Isabelle Marty
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
- * Correspondence should be adressed to: Isabelle Marty
| |
Collapse
|
46
|
Vassilopoulos S, Thevenon D, Rezgui SS, Brocard J, Chapel A, Lacampagne A, Lunardi J, De Waard M, Marty I. Triadins are not triad-specific proteins: two new skeletal muscle triadins possibly involved in the architecture of sarcoplasmic reticulum. J Biol Chem 2005; 280:28601-9. [PMID: 15927957 PMCID: PMC2739232 DOI: 10.1074/jbc.m501484200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We have cloned two new triadin isoforms from rat skeletal muscle, Trisk 49 and Trisk 32, which were named according to their theoretical molecular masses (49 and 32 kDa, respectively). Specific antibodies directed against each protein were produced to characterize both new triadins. Both are expressed in adult rat skeletal muscle, and their expression in slow twitch muscle is lower than that in fast twitch muscle. Using double immunofluorescent labeling, the localization of these two triadins was studied in comparison to well-characterized proteins such as ryanodine receptor, calsequestrin, desmin, Ca(2+)-ATPase, and titin. None of these two triadins are localized within the rat skeletal muscle triad. Both are instead found in different parts of the longitudinal sarcoplasmic reticulum. We attempted to identify partners for each isoform: neither is associated with ryanodine receptor; Trisk 49 could be associated with titin or another sarcomeric protein; and Trisk 32 could be associated with IP(3) receptor. These results open further fields of research concerning the functions of these two proteins; in particular, they could be involved in the set up and maintenance of a precise sarcoplasmic reticulum structure.
Collapse
Affiliation(s)
- Stéphane Vassilopoulos
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Dominique Thevenon
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Sophia Smida Rezgui
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Julie Brocard
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Agnès Chapel
- DRDC, Département réponse et dynamique cellulaire
INSERM : IFR27CNRS : IFR27INRACEAbat. C3 17 Rue des martyrs 38054 GRENOBLE CEDEX 9,FR
| | - Alain Lacampagne
- Physiopathologie cardiovasculaire
INSERM : U637IFR3Université Montpellier IHopital Arnaud de Villeneuve 171, Avenue du Doyen Gaston Giraud 34295 MONTPELLIER CEDEX 5,FR
- CHU Arnaud de Villeneuve
CHRU MontpellierMontpellier F-3400,,FR
| | - Joël Lunardi
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Michel De Waard
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Isabelle Marty
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
- * Correspondence should be adressed to: Isabelle Marty
| |
Collapse
|
47
|
Szappanos H, Smida-Rezgui S, Cseri J, Simut C, Sabatier JM, De Waard M, Kovács L, Csernoch L, Ronjat M. Differential effects of maurocalcine on Ca2+ release events and depolarization-induced Ca2+ release in rat skeletal muscle. J Physiol 2005; 565:843-53. [PMID: 15831537 PMCID: PMC1464547 DOI: 10.1113/jphysiol.2005.086074] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Maurocalcine (MCa), a 33 amino acid toxin obtained from scorpion venom, has been shown to interact with the isolated skeletal-type ryanodine receptor (RyR1) and to strongly modify its calcium channel gating. In this study, we explored the effects of MCa on RyR1 in situ to establish whether the functional interaction of RyR1 with the voltage-sensing dihydropyridine receptor (DHPR) would modify the ability of MCa to interact with RyR1. In developing skeletal muscle cells the addition of MCa into the external medium induced a calcium transient resulting from RyR1 activation and strongly inhibited the effect of the RyR1 agonist chloro-m-cresol. In contrast, MCa failed to affect the depolarization-induced Ca(2+) release. In intact adult fibres MCa did not induce any change in the cytosolic Ca(2+) concentration. However, when the surface membrane was permeabilized and calcium release events were readily observable, MCa had a time-dependent dual effect: it first increased event frequency, from 0.060 +/- 0.002 to 0.150 +/- 0.007 sarcomere(-1) s(-1), and reduced the amplitude of individual events without modifying their spatial distribution. Later on it induced the appearance of long-lasting events resembling the embers observed in control conditions but having a substantially longer duration. We propose that the functional coupling of DHPRs and RyR1s within a Ca(2+) release unit prevents MCa from either reaching its binding site or from being able to modify the gating not only of the RyR1s physically coupled to DHPRs but all RyR1s within the Ca(2+) release unit.
Collapse
|
48
|
Serysheva II. Structural insights into excitation-contraction coupling by electron cryomicroscopy. BIOCHEMISTRY (MOSCOW) 2005; 69:1226-32. [PMID: 15627376 DOI: 10.1007/s10541-005-0068-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In muscle, excitation-contraction coupling is defined as the process linking depolarization of the surface membrane with Ca2+ release from cytoplasmic stores, which activates contraction of striated muscle. This process is primarily controlled by interplay between two Ca2+ channels--the voltage-gated L-type Ca2+ channel (dihydropyridine receptor, DHPR) localized in the t-tubule membrane and the Ca2+-release channel (ryanodine receptor, RyR) of the sarcoplasmic reticulum membrane. The structures of both channels have been extensively studied by several groups using electron cryomicroscopy and single particle reconstruction techniques. The structures of RyR, determined at resolutions of 22-30 A, reveal a characteristic mushroom shape with a bulky cytoplasmic region and the membrane-spanning stem. While the cytoplasmic region exhibits a complex structure comprising a multitude of distinctive domains with numerous intervening cavities, at this resolution no definitive statement can be made about the location of the actual pore within the transmembrane region. Conformational changes associated with functional transitions of the Ca2+ release channel from closed to open states have been characterized. Further experiments determined localization of binding sites for various channel ligands. The structural studies of the DHPR are less developed. Although four 3D maps of the DHPR were reported recently at 24-30 A resolution from studies of frozen-hydrated and negatively stained receptors, there are some discrepancies between reported structures with respect to the overall appearance and dimensions of the channel structure. Future structural studies at higher resolution are needed to refine the structures of both channels and to substantiate a proposed molecular model for their interaction.
Collapse
Affiliation(s)
- I I Serysheva
- Department of Molecular Physiology and Biophysics, National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Darszon A, Nishigaki T, Wood C, Treviño CL, Felix R, Beltrán C. Calcium Channels and Ca2+ Fluctuations in Sperm Physiology. INTERNATIONAL REVIEW OF CYTOLOGY 2005; 243:79-172. [PMID: 15797459 DOI: 10.1016/s0074-7696(05)43002-8] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Generating new life in animals by sexual reproduction depends on adequate communication between mature and competent male and female gametes. Ion channels are instrumental in the dialogue between sperm, its environment, and the egg. The ability of sperm to swim to the egg and fertilize it is modulated by ion permeability changes induced by environmental cues and components of the egg outer layer. Ca(2+) is probably the key messenger in this information exchange. It is therefore not surprising that different Ca(2+)-permeable channels are distinctly localized in these tiny specialized cells. New approaches to measure sperm currents, intracellular Ca(2+), membrane potential, and intracellular pH with fluorescent probes, patch-clamp recordings, sequence information, and heterologous expression are revealing how sperm channels participate in fertilization. Certain sperm ion channels are turning out to be unique, making them attractive targets for contraception and for the discovery of novel signaling complexes.
Collapse
Affiliation(s)
- Alberto Darszon
- Department of Developmental Genetics and Molecular Physiology, Institute of Biotechnology, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico 62210
| | | | | | | | | | | |
Collapse
|
50
|
Altafaj X, Cheng W, Estève E, Urbani J, Grunwald D, Sabatier JM, Coronado R, De Waard M, Ronjat M. Maurocalcine and domain A of the II-III loop of the dihydropyridine receptor Cav 1.1 subunit share common binding sites on the skeletal ryanodine receptor. J Biol Chem 2004; 280:4013-6. [PMID: 15591063 PMCID: PMC2712624 DOI: 10.1074/jbc.c400433200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Maurocalcine is a scorpion venom toxin of 33 residues that bears a striking resemblance to the domain A of the dihydropyridine voltage-dependent calcium channel type 1.1 (Cav1.1) subunit. This domain belongs to the II-III loop of Cav1.1, which is implicated in excitation-contraction coupling. Besides the structural homology, maurocalcine also modulates RyR1 channel activity in a manner akin to a synthetic peptide of domain A. Because of these similarities, we hypothesized that maurocalcine and domain A may bind onto an identical region(s) of RyR1. Using a set of RyR1 fragments, we demonstrate that peptide A and maurocalcine bind onto two discrete RyR1 regions: fragments 3 and 7 encompassing residues 1021-1631 and 3201-3661, respectively. The binding onto fragment 7 is of greater importance and was thus further investigated. We found that the amino acid region 3351-3507 of RyR1 (fragment 7.2) is sufficient for these interactions. Proof that peptide A and maurocalcine bind onto the same site is provided by competition experiments in which binding of fragment 7.2 to peptide A is inhibited by preincubation with maurocalcine. Moreover, when expressed in COS-7 cells, RyR1 carrying a deletion of fragment 7 shows a loss of interaction with both peptide A and maurocalcine. At the functional level, this deletion abolishes the maurocalcine induced stimulation of [3H]ryanodine binding onto microsomes of transfected COS-7 cells without affecting the caffeine and ATP responses.
Collapse
Affiliation(s)
- Xavier Altafaj
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Weijun Cheng
- Department of Physiology
University of Wisconsin School of MedicineMadison, Wisconsin 53706,US
| | - Eric Estève
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Julie Urbani
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Didier Grunwald
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Jean-Marc Sabatier
- Biochimie - Ingénierie des protéines
CNRS : UMR6560Université de la Méditerranée - Aix-Marseille IIBoulevard Pierre Dramart 13916 Marseille Cedex 20,FR
| | - Roberto Coronado
- Department of Physiology
University of Wisconsin School of MedicineMadison, Wisconsin 53706,US
| | - Michel De Waard
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
| | - Michel Ronjat
- Canaux calciques , fonctions et pathologies
INSERM : U607CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble I17, rue des martyrs 38054 Grenoble,FR
- * Correspondence should be adressed to: Michel Ronjat
| |
Collapse
|