1
|
Tan RL, Sciandra F, Hübner W, Bozzi M, Reimann J, Schoch S, Brancaccio A, Blaess S. The missense mutation C667F in murine β-dystroglycan causes embryonic lethality, myopathy and blood-brain barrier destabilization. Dis Model Mech 2024; 17:dmm050594. [PMID: 38616731 PMCID: PMC11212641 DOI: 10.1242/dmm.050594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024] Open
Abstract
Dystroglycan (DG) is an extracellular matrix receptor consisting of an α- and a β-DG subunit encoded by the DAG1 gene. The homozygous mutation (c.2006G>T, p.Cys669Phe) in β-DG causes muscle-eye-brain disease with multicystic leukodystrophy in humans. In a mouse model of this primary dystroglycanopathy, approximately two-thirds of homozygous embryos fail to develop to term. Mutant mice that are born undergo a normal postnatal development but show a late-onset myopathy with partially penetrant histopathological changes and an impaired performance on an activity wheel. Their brains and eyes are structurally normal, but the localization of mutant β-DG is altered in the glial perivascular end-feet, resulting in a perturbed protein composition of the blood-brain and blood-retina barrier. In addition, α- and β-DG protein levels are significantly reduced in muscle and brain of mutant mice. Owing to the partially penetrant developmental phenotype of the C669F β-DG mice, they represent a novel and highly valuable mouse model with which to study the molecular effects of β-DG functional alterations both during embryogenesis and in mature muscle, brain and eye, and to gain insight into the pathogenesis of primary dystroglycanopathies.
Collapse
Affiliation(s)
- Rui Lois Tan
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Francesca Sciandra
- Institute of Chemical Sciences and Technologies 'Giulio Natta' (SCITEC)-CNR, 00168 Rome, Italy
| | - Wolfgang Hübner
- Biomolecular Photonics, Faculty of Physics, Bielefeld University, 33615 Bielefeld, Germany
| | - Manuela Bozzi
- Institute of Chemical Sciences and Technologies 'Giulio Natta' (SCITEC)-CNR, 00168 Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie. Sezione di Biochimica. Università Cattolica del Sacro Cuore di Roma, 00168 Rome, Italy
| | - Jens Reimann
- Department of Neurology, Neuromuscular Diseases Section, University Hospital Bonn, 53127 Bonn, Germany
| | - Susanne Schoch
- Synaptic Neuroscience Team, Institute of Neuropathology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Andrea Brancaccio
- Institute of Chemical Sciences and Technologies 'Giulio Natta' (SCITEC)-CNR, 00168 Rome, Italy
- School of Biochemistry, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
2
|
Benucci S, Ruiz A, Franchini M, Ruggiero L, Zoppi D, Sitsapesan R, Lindsay C, Pelczar P, Pietrangelo L, Protasi F, Treves S, Zorzato F. A novel, patient-derived RyR1 mutation impairs muscle function and calcium homeostasis in mice. J Gen Physiol 2024; 156:e202313486. [PMID: 38445312 PMCID: PMC10911087 DOI: 10.1085/jgp.202313486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/20/2023] [Accepted: 02/09/2024] [Indexed: 03/07/2024] Open
Abstract
RYR1 is the most commonly mutated gene associated with congenital myopathies, a group of early-onset neuromuscular conditions of variable severity. The functional effects of a number of dominant RYR1 mutations have been established; however, for recessive mutations, these effects may depend on multiple factors, such as the formation of a hypomorphic allele, or on whether they are homozygous or compound heterozygous. Here, we functionally characterize a new transgenic mouse model knocked-in for mutations identified in a severely affected child born preterm and presenting limited limb movement. The child carried the homozygous c.14928C>G RYR1 mutation, resulting in the p.F4976L substitution. In vivo and ex vivo assays revealed that homozygous mice fatigued sooner and their muscles generated significantly less force compared with their WT or heterozygous littermates. Electron microscopy, biochemical, and physiological analyses showed that muscles from RyR1 p.F4976L homozygous mice have the following properties: (1) contain fewer calcium release units and show areas of myofibrillar degeneration, (2) contain less RyR1 protein, (3) fibers show smaller electrically evoked calcium transients, and (4) their SR has smaller calcium stores. In addition, single-channel recordings indicate that RyR1 p.F4976L exhibits higher Po in the presence of 100 μM [Ca2+]. Our mouse model partly recapitulates the clinical picture of the homozygous human patient and provides significant insight into the functional impact of this mutation. These results will help understand the pathology of patients with similar RYR1 mutations.
Collapse
Affiliation(s)
- Sofia Benucci
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Alexis Ruiz
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Martina Franchini
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
| | - Lucia Ruggiero
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Dario Zoppi
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | | | - Chris Lindsay
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Laura Pietrangelo
- DMSI, Department of Medicine and Aging Sciences and CAST, Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- DMSI, Department of Medicine and Aging Sciences and CAST, Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Susan Treves
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesco Zorzato
- Departments of Biomedicine and Neurology, Basel University Hospital, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
3
|
Marcucci L, Michelucci A, Reggiani C. Cytosolic Ca 2+ gradients and mitochondrial Ca 2+ uptake in resting muscle fibers: A model analysis. BIOPHYSICAL REPORTS 2023; 3:100117. [PMID: 37576797 PMCID: PMC10412765 DOI: 10.1016/j.bpr.2023.100117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
Calcium ions (Ca2+) enter mitochondria via the mitochondrial Ca2+ uniporter, driven by electrical and concentration gradients. In this regard, transgenic mouse models, such as calsequestrin knockout (CSQ-KO) mice, with higher mitochondrial Ca2+ concentrations ([Ca2+]mito), should display higher cytosolic Ca2+ concentrations ([Ca2+]cyto). However, repeated measurements of [Ca2+]cyto in quiescent CSQ-KO fibers never showed a difference between WT and CSQ-KO. Starting from the consideration that fluorescent Ca2+ probes (Fura-2 and Indo-1) measure averaged global cytosolic concentrations, in this report we explored the role of local Ca2+ concentrations (i.e., Ca2+ microdomains) in regulating mitochondrial Ca2+ in resting cells, using a multicompartmental diffusional Ca2+ model. Progressively including the inward and outward fluxes of sarcoplasmic reticulum (SR), extracellular space, and mitochondria, we explored their contribution to the local Ca2+ distribution within the cell. The model predicts Ca2+ concentration gradients with hot spots or microdomains even at rest, minor but similar to those of evoked Ca2+ release. Due to their specific localization close to Ca2+ release units (CRU), mitochondria could take up Ca2+ directly from high-concentration microdomains, thus sensibly raising [Ca2+]mito, despite minor, possibly undetectable, modifications of the average [Ca2+]cyto.
Collapse
Affiliation(s)
- Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Center for Biosystems Dynamics Research, RIKEN, Suita, Japan
| | - Antonio Michelucci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, Koper, Slovenia
| |
Collapse
|
4
|
Ruiz A, Benucci S, Duthaler U, Bachmann C, Franchini M, Noreen F, Pietrangelo L, Protasi F, Treves S, Zorzato F. Improvement of muscle strength in a mouse model for congenital myopathy treated with HDAC and DNA methyltransferase inhibitors. eLife 2022; 11:73718. [PMID: 35238775 PMCID: PMC8956288 DOI: 10.7554/elife.73718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
To date there are no therapies for patients with congenital myopathies, muscle disorders causing poor quality of life of affected individuals. In approximately 30% of the cases, patients with congenital myopathies carry either dominant or recessive mutations in the RYR1 gene; recessive RYR1 mutations are accompanied by reduction of RyR1 expression and content in skeletal muscles and are associated with fiber hypotrophy and muscle weakness. Importantly, muscles of patients with recessive RYR1 mutations exhibit increased content of class II histone de-acetylases and of DNA genomic methylation. We recently created a mouse model knocked-in for the p.Q1970fsX16+p.A4329D RyR1 mutations, which are isogenic to those carried by a severely affected child suffering from a recessive form of RyR1-related multi-mini core disease. The phenotype of the RyR1 mutant mice recapitulates many aspects of the clinical picture of patients carrying recessive RYR1 mutations. We treated the compound heterozygous mice with a combination of two drugs targeting DNA methylases and class II histone de-acetylases. Here we show that treatment of the mutant mice with drugs targeting epigenetic enzymes improves muscle strength, RyR1 protein content and muscle ultrastructure. This study provides proof of concept for the pharmacological treatment of patients with congenital myopathies linked to recessive RYR1 mutations.
Collapse
Affiliation(s)
- Alexis Ruiz
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Sofia Benucci
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Urs Duthaler
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Christoph Bachmann
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Martina Franchini
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Faiza Noreen
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Laura Pietrangelo
- Department of Neuroscience, Imaging and Clinical Science, University G d' Annunzio of Chieti, Chieti, Italy
| | - Feliciano Protasi
- Department of Neuroscience, Imaging and Clinical Science, University G d' Annunzio of Chieti, Chieti, Italy
| | - Susan Treves
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Francesco Zorzato
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
5
|
Jaque-Fernández F, Jorquera G, Troc-Gajardo J, Pietri-Rouxel F, Gentil C, Buvinic S, Allard B, Jaimovich E, Jacquemond V, Casas M. Pannexin-1 and CaV1.1 show reciprocal interaction during excitation-contraction and excitation-transcription coupling in skeletal muscle. J Gen Physiol 2021; 153:212695. [PMID: 34636893 PMCID: PMC8515650 DOI: 10.1085/jgp.202012635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/24/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023] Open
Abstract
One of the most important functions of skeletal muscle is to respond to nerve stimuli by contracting. This function ensures body movement but also participates in other important physiological roles, like regulation of glucose homeostasis. Muscle activity is closely regulated to adapt to different demands and shows a plasticity that relies on both transcriptional activity and nerve stimuli. These two processes, both dependent on depolarization of the plasma membrane, have so far been regarded as separated and independent processes due to a lack of evidence of common protein partners or molecular mechanisms. In this study, we reveal intimate functional interactions between the process of excitation-induced contraction and the process of excitation-induced transcriptional activity in skeletal muscle. We show that the plasma membrane voltage-sensing protein CaV1.1 and the ATP-releasing channel Pannexin-1 (Panx1) regulate each other in a reciprocal manner, playing roles in both processes. Specifically, knockdown of CaV1.1 produces chronically elevated extracellular ATP concentrations at rest, consistent with disruption of the normal control of Panx1 activity. Conversely, knockdown of Panx1 affects not only activation of transcription but also CaV1.1 function on the control of muscle fiber contraction. Altogether, our results establish the presence of bidirectional functional regulations between the molecular machineries involved in the control of contraction and transcription induced by membrane depolarization of adult muscle fibers. Our results are important for an integrative understanding of skeletal muscle function and may impact our understanding of several neuromuscular diseases.
Collapse
Affiliation(s)
- Francisco Jaque-Fernández
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Gonzalo Jorquera
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jennifer Troc-Gajardo
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - France Pietri-Rouxel
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Christel Gentil
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Enrique Jaimovich
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Mariana Casas
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Michelucci A, Boncompagni S, Pietrangelo L, Takano T, Protasi F, Dirksen RT. Pre-assembled Ca2+ entry units and constitutively active Ca2+ entry in skeletal muscle of calsequestrin-1 knockout mice. J Gen Physiol 2021; 152:152001. [PMID: 32761048 PMCID: PMC7537346 DOI: 10.1085/jgp.202012617] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ influx mechanism triggered by depletion of Ca2+ stores from the endoplasmic/sarcoplasmic reticulum (ER/SR). We recently reported that acute exercise in WT mice drives the formation of Ca2+ entry units (CEUs), intracellular junctions that contain STIM1 and Orai1, the two key proteins mediating SOCE. The presence of CEUs correlates with increased constitutive- and store-operated Ca2+ entry, as well as sustained Ca2+ release and force generation during repetitive stimulation. Skeletal muscle from mice lacking calsequestrin-1 (CASQ1-null), the primary Ca2+-binding protein in the lumen of SR terminal cisternae, exhibits significantly reduced total Ca2+ store content and marked SR Ca2+ depletion during high-frequency stimulation. Here, we report that CEUs are constitutively assembled in extensor digitorum longus (EDL) and flexor digitorum brevis (FDB) muscles of sedentary CASQ1-null mice. The higher density of CEUs in EDL (39.6 ± 2.1/100 µm2 versus 2.0 ± 0.3/100 µm2) and FDB (16.7 ± 1.0/100 µm2 versus 2.7 ± 0.5/100 µm2) muscles of CASQ1-null compared with WT mice correlated with enhanced constitutive- and store-operated Ca2+ entry and increased expression of STIM1, Orai1, and SERCA. The higher ability to recover Ca2+ ions via SOCE in CASQ1-null muscle served to promote enhanced maintenance of peak Ca2+ transient amplitude, increased dependence of luminal SR Ca2+ replenishment on BTP-2-sensitive SOCE, and increased maintenance of contractile force during repetitive, high-frequency stimulation. Together, these data suggest that muscles from CASQ1-null mice compensate for the lack of CASQ1 and reduction in total releasable SR Ca2+ content by assembling CEUs to promote constitutive and store-operated Ca2+ entry.
Collapse
Affiliation(s)
- Antonio Michelucci
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY.,Center for Advanced Studies and Technologies, University G. d'Annunzio of Chieti, Chieti, Italy
| | - Simona Boncompagni
- Center for Advanced Studies and Technologies, University G. d'Annunzio of Chieti, Chieti, Italy.,Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti, Chieti, Italy
| | - Laura Pietrangelo
- Center for Advanced Studies and Technologies, University G. d'Annunzio of Chieti, Chieti, Italy.,Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti, Chieti, Italy
| | - Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Feliciano Protasi
- Center for Advanced Studies and Technologies, University G. d'Annunzio of Chieti, Chieti, Italy.,Department of Medicine and Ageing Sciences, University G. d'Annunzio of Chieti, Chieti, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
7
|
Reddish FN, Miller CL, Deng X, Dong B, Patel AA, Ghane MA, Mosca B, McBean C, Wu S, Solntsev KM, Zhuo Y, Gadda G, Fang N, Cox DN, Mabb AM, Treves S, Zorzato F, Yang JJ. Rapid subcellular calcium responses and dynamics by calcium sensor G-CatchER . iScience 2021; 24:102129. [PMID: 33665552 PMCID: PMC7900224 DOI: 10.1016/j.isci.2021.102129] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 12/14/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
The precise spatiotemporal characteristics of subcellular calcium (Ca2+) transients are critical for the physiological processes. Here we report a green Ca2+ sensor called "G-CatchER+" using a protein design to report rapid local ER Ca2+ dynamics with significantly improved folding properties. G-CatchER+ exhibits a superior Ca2+ on rate to G-CEPIA1er and has a Ca2+-induced fluorescence lifetimes increase. G-CatchER+ also reports agonist/antagonist triggered Ca2+ dynamics in several cell types including primary neurons that are orchestrated by IP3Rs, RyRs, and SERCAs with an ability to differentiate expression. Upon localization to the lumen of the RyR channel (G-CatchER+-JP45), we report a rapid local Ca2+ release that is likely due to calsequestrin. Transgenic expression of G-CatchER+ in Drosophila muscle demonstrates its utility as an in vivo reporter of stimulus-evoked SR local Ca2+ dynamics. G-CatchER+ will be an invaluable tool to examine local ER/SR Ca2+ dynamics and facilitate drug development associated with ER dysfunction.
Collapse
Affiliation(s)
- Florence N. Reddish
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Cassandra L. Miller
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaonan Deng
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Bin Dong
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Atit A. Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Mohammad A. Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Barbara Mosca
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
| | - Cheyenne McBean
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Shengnan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Kyril M. Solntsev
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - You Zhuo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Giovanni Gadda
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Ning Fang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Susan Treves
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
- Department of Biomedicine, Basel University, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Francesco Zorzato
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
- Department of Biomedicine, Basel University, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Jenny J. Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
8
|
Kuhn B, Picollo F, Carabelli V, Rispoli G. Advanced real-time recordings of neuronal activity with tailored patch pipettes, diamond multi-electrode arrays and electrochromic voltage-sensitive dyes. Pflugers Arch 2020; 473:15-36. [PMID: 33047171 PMCID: PMC7782438 DOI: 10.1007/s00424-020-02472-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/03/2022]
Abstract
To understand the working principles of the nervous system is key to figure out its electrical activity and how this activity spreads along the neuronal network. It is therefore crucial to develop advanced techniques aimed to record in real time the electrical activity, from compartments of single neurons to populations of neurons, to understand how higher functions emerge from coordinated activity. To record from single neurons, a technique will be presented to fabricate patch pipettes able to seal on any membrane with a single glass type and whose shanks can be widened as desired. This dramatically reduces access resistance during whole-cell recording allowing fast intracellular and, if required, extracellular perfusion. To simultaneously record from many neurons, biocompatible probes will be described employing multi-electrodes made with novel technologies, based on diamond substrates. These probes also allow to synchronously record exocytosis and neuronal excitability and to stimulate neurons. Finally, to achieve even higher spatial resolution, it will be shown how voltage imaging, employing fast voltage-sensitive dyes and two-photon microscopy, is able to sample voltage oscillations in the brain spatially resolved and voltage changes in dendrites of single neurons at millisecond and micrometre resolution in awake animals.
Collapse
Affiliation(s)
- Bernd Kuhn
- Optical Neuroimaging Unit, OIST Graduate University, 1919-1 Tancha, Onna-son, Okinawa, Japan
| | - Federico Picollo
- Department of Physics, NIS Interdepartmental Centre, University of Torino and Italian Institute of Nuclear Physics, via Giuria 1, 10125, Torino, Italy
| | - Valentina Carabelli
- Department of Drug and Science Technology, NIS Interdepartmental Centre, University of Torino, Corso Raffaello 30, 10125, Torino, Italy
| | - Giorgio Rispoli
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy.
| |
Collapse
|
9
|
Li P, Cai X, Xiao N, Ma X, Zeng L, Zhang LH, Xie L, Du B. Sacha inchi ( Plukenetia volubilis L.) shell extract alleviates hypertension in association with the regulation of gut microbiota. Food Funct 2020; 11:8051-8067. [PMID: 32852030 DOI: 10.1039/d0fo01770a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dysbiosis of gut microbiota has been implicated in the pathogenesis of hypertension. A definite relationship between gut microbiota and hypertension remains intriguing. Here, we show that the Sacha inchi (Plukenetia volubilis L.) shell extract (SISE) intervention significantly reduced systolic blood pressures in spontaneous hypertensive rats (SHR), attenuated the oxidative damage and modulated plasma calcium homeostasis and left ventricular hypertrophy in both SHR and high-salt diet Wistar-Kyoto rats. SISE reshaped the gut microbiome and metabolome, particularly by improving the prevalence of Roseburia and dihydrofolic acid levels in the gut. Transcriptome analyses showed that the protective effects of SISE were accompanied by the modulation of renal molecular pathways, beneficial for cardiovascular functions such as the L-type voltage-dependent calcium channel (LTCC), a key regulator of calcium signaling. Overall, the results have shown that dietary SISE can alleviate hypertension regulating the gut microbiota, and Ca2+ signaling might be a potential target for spontaneous hypertension.
Collapse
Affiliation(s)
- Pan Li
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xin Cai
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Nan Xiao
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaowei Ma
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Liping Zeng
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Lian-Hui Zhang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou 510642, China.
| | - Lanhua Xie
- Expert Research Station of Bing Du, Pu'er City, Yunnan 665000, China.
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou 510642, China and Expert Research Station of Bing Du, Pu'er City, Yunnan 665000, China.
| |
Collapse
|
10
|
Elbaz M, Ruiz A, Nicolay S, Tupini C, Bachmann C, Eckhardt J, Benucci S, Pelczar P, Treves S, Zorzato F. Bi-allelic expression of the RyR1 p.A4329D mutation decreases muscle strength in slow-twitch muscles in mice. J Biol Chem 2020; 295:10331-10339. [PMID: 32499372 DOI: 10.1074/jbc.ra120.013846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/29/2020] [Indexed: 12/25/2022] Open
Abstract
Mutations in the ryanodine receptor 1 (RYR1) gene are associated with several human congenital myopathies, including the dominantly inherited central core disease and exercise-induced rhabdomyolysis, and the more severe recessive phenotypes, including multiminicore disease, centronuclear myopathy, and congenital fiber type disproportion. Within the latter group, those carrying a hypomorphic mutation in one allele and a missense mutation in the other are the most severely affected. Because of nonsense-mediated decay, most hypomorphic alleles are not expressed, resulting in homozygous expression of the missense mutation allele. This should result in 50% reduced expression of the ryanodine receptor in skeletal muscle, but its observed content is even lower. To study in more detail the biochemistry and pathophysiology of recessive RYR1 myopathies, here we investigated a mouse model we recently generated by analyzing the effect of bi-allelic versus mono-allelic expression of the RyR1 p.A4329D mutation. Our results revealed that the expression of two alleles carrying the same mutation or of one allele with the mutation in combination with a hypomorphic allele does not result in functionally equal outcomes and impacts skeletal muscles differently. In particular, the bi-allelic RyR1 p.A4329D mutation caused a milder phenotype than its mono-allelic expression, leading to changes in the biochemical properties and physiological function only of slow-twitch muscles and largely sparing fast-twitch muscles. In summary, bi-allelic expression of the RyR1 p.A4329D mutation phenotypically differs from mono-allelic expression of this mutation in a compound heterozygous carrier.
Collapse
Affiliation(s)
- Moran Elbaz
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Alexis Ruiz
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Sven Nicolay
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Chiara Tupini
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Christoph Bachmann
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Jan Eckhardt
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Sofia Benucci
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Susan Treves
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland.,Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesco Zorzato
- Department of Biomedicine, Basel University Hospital, Basel, Switzerland .,Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
11
|
Elbaz M, Ruiz A, Bachmann C, Eckhardt J, Pelczar P, Venturi E, Lindsay C, Wilson AD, Alhussni A, Humberstone T, Pietrangelo L, Boncompagni S, Sitsapesan R, Treves S, Zorzato F. Quantitative RyR1 reduction and loss of calcium sensitivity of RyR1Q1970fsX16+A4329D cause cores and loss of muscle strength. Hum Mol Genet 2020; 28:2987-2999. [PMID: 31044239 DOI: 10.1093/hmg/ddz092] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/25/2019] [Accepted: 04/25/2019] [Indexed: 01/10/2023] Open
Abstract
Recessive ryanodine receptor 1 (RYR1) mutations cause congenital myopathies including multiminicore disease (MmD), congenital fiber-type disproportion and centronuclear myopathy. We created a mouse model knocked-in for the Q1970fsX16+A4329D RYR1 mutations, which are isogenic with those identified in a severely affected child with MmD. During the first 20 weeks after birth the body weight and the spontaneous running distance of the mutant mice were 20% and 50% lower compared to wild-type littermates. Skeletal muscles from mutant mice contained 'cores' characterized by severe myofibrillar disorganization associated with misplacement of mitochondria. Furthermore, their muscles developed less force and had smaller electrically evoked calcium transients. Mutant RyR1 channels incorporated into lipid bilayers were less sensitive to calcium and caffeine, but no change in single-channel conductance was observed. Our results demonstrate that the phenotype of the RyR1Q1970fsX16+A4329D compound heterozygous mice recapitulates the clinical picture of multiminicore patients and provide evidence of the molecular mechanisms responsible for skeletal muscle defects.
Collapse
Affiliation(s)
- Moran Elbaz
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Alexis Ruiz
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Christoph Bachmann
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Jan Eckhardt
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Mattenstrasse 22, 4002 Basel, Switzerland
| | - Elisa Venturi
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Chris Lindsay
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.,Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford OX1 3TA, UK
| | - Abigail D Wilson
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Ahmed Alhussni
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Thomas Humberstone
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Laura Pietrangelo
- Center for Research on Ageing and Translational Medicine and Department of Neuroscience, Imaging and Clinical Sciences, Università G. d'Annunzio, 66100 Chieti, Italy
| | - Simona Boncompagni
- Center for Research on Ageing and Translational Medicine and Department of Neuroscience, Imaging and Clinical Sciences, Università G. d'Annunzio, 66100 Chieti, Italy
| | - Rebecca Sitsapesan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Susan Treves
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, 44100, Ferrara, Italy
| | - Francesco Zorzato
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, 44100, Ferrara, Italy
| |
Collapse
|
12
|
Beqollari D, Kohrt WM, Bannister RA. Equivalent L-type channel (Ca V1.1) function in adult female and male mouse skeletal muscle fibers. Biochem Biophys Res Commun 2019; 522:996-1002. [PMID: 31812241 DOI: 10.1016/j.bbrc.2019.11.164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 11/16/2022]
Abstract
Loss of total muscle force during aging has both atrophic and non-atrophic components. The former deficit is a direct consequence of reduced muscle mass while the latter has been attributed to a depression of excitation-contraction (EC) coupling. It is well established that age-onset reductions in sex hormone production regulate the atrophic component in both males and females. However, it is unknown whether the non-atrophic component is influenced by sex hormones. Since the non-atrophic component has been linked mechanistically to reduced expression of the skeletal muscle L-type Ca2+ channel (CaV1.1), we recorded L-type Ca2+ currents, gating charge movements and depolarization-induced changes in myoplasmic Ca2+ from flexor digitorum brevis (FDB) fibers of naïve and gonadectomized mice of both sexes. Our first set of experiments sought to identify any basal differences in EC coupling or L-type Ca2+ flux between the sexes; no detectable differences in any of the aforementioned parameters were observed between FDB harvested from either naïve males or females. In the latter segments of the study, ovariectomy (OVX) and orchiectomy (ORX) models were used to assess the possible influence of sex hormones on EC coupling and/or L-type Ca2+ flux. In these experiments, FDB fibers harvested from OVX and ORX mice both showed no differences in L-type Ca2+ current, gating charge movement or depolarization-induced changes in Ca2+ release from the sarcoplasmic reticulum. Taken together, our results indicate L-type Ca2+ channel function and EC coupling are: 1) equivalent between the sexes, and 2) not significantly regulated by sex hormones. Since recent NIH review guidelines mandate the consideration of sex differences as a criterion for review, our work indicates the suitability of either sex for the study of the fundamental mechanisms of EC coupling. Thus, our findings may accelerate the research process by conserving animals, labor and financial resources.
Collapse
Affiliation(s)
- D Beqollari
- Department of Medicine - Division of Cardiology, University of Colorado School of Medicine, 12800 East 19th Avenue, P15-8006, Box 139, Aurora, CO, 80045, USA.
| | - W M Kohrt
- Department of Medicine - Division of Geriatric Medicine, University of Colorado School of Medicine, 12631 East 17th Avenue, L15-8000, Aurora, CO, 80045, USA.
| | - R A Bannister
- Department of Medicine - Division of Cardiology, University of Colorado School of Medicine, 12800 East 19th Avenue, P15-8006, Box 139, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Elbaz M, Ruiz A, Eckhardt J, Pelczar P, Muntoni F, Boncompagni S, Treves S, Zorzato F. Quantitative reduction of RyR1 protein caused by a single-allele frameshift mutation in RYR1 ex36 impairs the strength of adult skeletal muscle fibres. Hum Mol Genet 2019; 28:1872-1884. [DOI: 10.1093/hmg/ddz025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Moran Elbaz
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse, Basel, Switzerland
| | - Alexis Ruiz
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse, Basel, Switzerland
| | - Jan Eckhardt
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse, Basel, Switzerland
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Mattenstrasse, Basel, Switzerland
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Simona Boncompagni
- Center for Research on Ageing and Translational Medicine & DNICS - Deptartment of Neuroscience, Imaging and Clinical Sciences, Università degli Studi G. d'Annunzio, Chieti, Italy
| | - Susan Treves
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Via Borsari, Ferrara, Italy
| | - Francesco Zorzato
- Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse, Basel, Switzerland
- Department of Life Science and Biotechnology, University of Ferrara, Via Borsari, Ferrara, Italy
| |
Collapse
|
14
|
Abstract
Cacna1s encodes the α1S subunit (Cav1.1) of voltage-dependent calcium channels, and is required for normal skeletal and cardiac muscle function, where it couples with the ryanodine receptor to regulate muscle contraction. Recently CACNA1S was reported to be expressed on the tips of retinal depolarizing bipolar cells (DBCs) and colocalized with metabotropic glutamate receptor 6 (mGluR6), which is critical to DBC signal transduction. Further, in mGluR6 knockout mice, expression at this location is down regulated. We examined RNAseq data from mouse retina and found expression of a novel isoform of Cacna1s. To determine if CACNA1S was a functional component of the DBC signal transduction cascade, we performed immunohistochemistry to visualize its expression in several mouse lines that lack DBC function. Immunohistochemical staining with antibodies to CACNA1S show punctate labeling at the tips of DBCs in wild type (WT) retinas that are absent in Gpr179 nob5 mutant retinas and decreased in Grm6 -/- mouse retinas. CACNA1S and transient receptor potential cation channel, subfamily M, member 1 (TRPM1) staining also colocalized in WT retinas. Western blot analyses for CACNA1S of either retinal lysates or proteins after immunoprecipitation with the CACNA1S antibody failed to show the presence of bands expected for CACNA1S. Mass spectrometric analysis of CACNA1S immunoprecipitated proteins also failed to detect any peptides matching CACNA1S. Immunohistochemistry and western blotting after expression of GPR179 in HEK293T cells indicate that the CACNA1S antibody used here and in the retinal studies published to date, cross-reacts with GPR179. These data suggest caution should be exercised in conferring a role for CACNA1S in DBC signal transduction based solely on immunohistochemical staining.
Collapse
|
15
|
Bannister RA, Sheridan DC, Beam KG. Distinct Components of Retrograde Ca(V)1.1-RyR1 Coupling Revealed by a Lethal Mutation in RyR1. Biophys J 2016; 110:912-21. [PMID: 26910427 DOI: 10.1016/j.bpj.2015.12.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/24/2015] [Accepted: 12/30/2015] [Indexed: 12/21/2022] Open
Abstract
The molecular basis for excitation-contraction coupling in skeletal muscle is generally thought to involve conformational coupling between the L-type voltage-gated Ca(2+) channel (CaV1.1) and the type 1 ryanodine receptor (RyR1). This coupling is bidirectional; in addition to the orthograde signal from CaV1.1 to RyR1 that triggers Ca(2+) release from the sarcoplasmic reticulum, retrograde signaling from RyR1 to CaV1.1 results in increased amplitude and slowed activation kinetics of macroscopic L-type Ca(2+) current. Orthograde coupling was previously shown to be ablated by a glycine for glutamate substitution at RyR1 position 4242. In this study, we investigated whether the RyR1-E4242G mutation affects retrograde coupling. L-type current in myotubes homozygous for RyR1-E4242G was substantially reduced in amplitude (∼80%) relative to that observed in myotubes from normal control (wild-type and/or heterozygous) myotubes. Analysis of intramembrane gating charge movements and ionic tail current amplitudes indicated that the reduction in current amplitude during step depolarizations was a consequence of both decreased CaV1.1 membrane expression (∼50%) and reduced channel Po (∼55%). In contrast, activation kinetics of the L-type current in RyR1-E4242G myotubes resembled those of normal myotubes, unlike dyspedic (RyR1 null) myotubes in which the L-type currents have markedly accelerated activation kinetics. Exogenous expression of wild-type RyR1 partially restored L-type current density. From these observations, we conclude that mutating residue E4242 affects RyR1 structures critical for retrograde communication with CaV1.1. Moreover, we propose that retrograde coupling has two distinct and separable components that are dependent on different structural elements of RyR1.
Collapse
Affiliation(s)
- Roger A Bannister
- Cardiology Division, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - David C Sheridan
- Department of Biology and Earth Science, Otterbein University, Westerville, Ohio
| | - Kurt G Beam
- Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
16
|
Beqollari D, Romberg CF, Dobrowolny G, Martini M, Voss AA, Musarò A, Bannister RA. Progressive impairment of CaV1.1 function in the skeletal muscle of mice expressing a mutant type 1 Cu/Zn superoxide dismutase (G93A) linked to amyotrophic lateral sclerosis. Skelet Muscle 2016; 6:24. [PMID: 27340545 PMCID: PMC4918102 DOI: 10.1186/s13395-016-0094-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 06/03/2016] [Indexed: 11/24/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disorder that is typically fatal within 3–5 years of diagnosis. While motoneuron death is the defining characteristic of ALS, the events that underlie its pathology are not restricted to the nervous system. In this regard, ALS muscle atrophies and weakens significantly before presentation of neurological symptoms. Since the skeletal muscle L-type Ca2+ channel (CaV1.1) is a key regulator of both mass and force, we investigated whether CaV1.1 function is impaired in the muscle of two distinct mouse models carrying an ALS-linked mutation. Methods We recorded L-type currents, charge movements, and myoplasmic Ca2+ transients from dissociated flexor digitorum brevis (FDB) fibers to assess CaV1.1 function in two mouse models expressing a type 1 Cu/Zn superoxide dismutase mutant (SOD1G93A). Results In FDB fibers obtained from “symptomatic” global SOD1G93A mice, we observed a substantial reduction of SR Ca2+ release in response to depolarization relative to fibers harvested from age-matched control mice. L-type current and charge movement were both reduced by ~40 % in symptomatic SOD1G93A fibers when compared to control fibers. Ca2+ transients were not significantly reduced in similar experiments performed with FDB fibers obtained from “early-symptomatic” SOD1G93A mice, but L-type current and charge movement were decreased (~30 and ~20 %, respectively). Reductions in SR Ca2+ release (~35 %), L-type current (~20 %), and charge movement (~15 %) were also observed in fibers obtained from another model where SOD1G93A expression was restricted to skeletal muscle. Conclusions We report reductions in EC coupling, L-type current density, and charge movement in FDB fibers obtained from symptomatic global SOD1G93A mice. Experiments performed with FDB fibers obtained from early-symptomatic SOD1G93A and skeletal muscle autonomous MLC/SOD1G93A mice support the idea that events occurring locally in the skeletal muscle contribute to the impairment of CaV1.1 function in ALS muscle independently of innervation status. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0094-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Donald Beqollari
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, 12700 East 19th Avenue, B-139, Aurora, CO 80045 USA
| | - Christin F Romberg
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, 12700 East 19th Avenue, B-139, Aurora, CO 80045 USA
| | - Gabriella Dobrowolny
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, La Sapienza University, Via A. Scarpa, 14, 00161 Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Martina Martini
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, La Sapienza University, Via A. Scarpa, 14, 00161 Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Andrew A Voss
- Department of Biological Sciences, College of Science and Mathematics, Wright State University, 235A Biological Sciences, 3640 Colonel Glenn Highway, Dayton, OH 45435 USA
| | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, La Sapienza University, Via A. Scarpa, 14, 00161 Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Roger A Bannister
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, 12700 East 19th Avenue, B-139, Aurora, CO 80045 USA
| |
Collapse
|
17
|
Mosca B, Eckhardt J, Bergamelli L, Treves S, Bongianino R, De Negri M, Priori SG, Protasi F, Zorzato F. Role of the JP45-Calsequestrin Complex on Calcium Entry in Slow Twitch Skeletal Muscles. J Biol Chem 2016; 291:14555-65. [PMID: 27189940 PMCID: PMC4938177 DOI: 10.1074/jbc.m115.709071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 12/27/2022] Open
Abstract
We exploited a variety of mouse models to assess the roles of JP45-CASQ1 (CASQ, calsequestrin) and JP45-CASQ2 on calcium entry in slow twitch muscles. In flexor digitorum brevis (FDB) fibers isolated from JP45-CASQ1-CASQ2 triple KO mice, calcium transients induced by tetanic stimulation rely on calcium entry via La3+- and nifedipine-sensitive calcium channels. The comparison of excitation-coupled calcium entry (ECCE) between FDB fibers from WT, JP45KO, CASQ1KO, CASQ2KO, JP45-CASQ1 double KO, JP45-CASQ2 double KO, and JP45-CASQ1-CASQ2 triple KO shows that ECCE enhancement requires ablation of both CASQs and JP45. Calcium entry activated by ablation of both JP45-CASQ1 and JP45-CASQ2 complexes supports tetanic force development in slow twitch soleus muscles. In addition, we show that CASQs interact with JP45 at Ca2+ concentrations similar to those present in the lumen of the sarcoplasmic reticulum at rest, whereas Ca2+ concentrations similar to those present in the SR lumen after depolarization-induced calcium release cause the dissociation of JP45 from CASQs. Our results show that the complex JP45-CASQs is a negative regulator of ECCE and that tetanic force development in slow twitch muscles is supported by the dynamic interaction between JP45 and CASQs.
Collapse
Affiliation(s)
- Barbara Mosca
- Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, 44100, Ferrara, Italy
| | - Jan Eckhardt
- From the Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Leda Bergamelli
- Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, 44100, Ferrara, Italy
| | - Susan Treves
- Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, 44100, Ferrara, Italy From the Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Rossana Bongianino
- Molecular Cardiology Laboratories Fondazione Salvatore Maugeri, Via Maugeri 10/10°, 27100, Pavia Italy
| | - Marco De Negri
- Molecular Cardiology Laboratories Fondazione Salvatore Maugeri, Via Maugeri 10/10°, 27100, Pavia Italy
| | - Silvia G Priori
- Molecular Cardiology Laboratories Fondazione Salvatore Maugeri, Via Maugeri 10/10°, 27100, Pavia Italy, Department of Molecular Medicine, University of Pavia, Pavia Italy, and
| | - Feliciano Protasi
- Center for Research on Ageing and Translational Medicine and DNICS (Department of Neuroscience, Imaging, and Clinical Sciences), University G. d'Annunzio, 66100 Chieti, Italy
| | - Francesco Zorzato
- From the Departments of Anaesthesia and Biomedicine, Basel University Hospital, Hebelstrasse 20, 4031 Basel, Switzerland, Department of Life Science and Biotechnology, University of Ferrara, Via Borsari 46, 44100, Ferrara, Italy
| |
Collapse
|
18
|
Ríos E, Figueroa L, Manno C, Kraeva N, Riazi S. The couplonopathies: A comparative approach to a class of diseases of skeletal and cardiac muscle. ACTA ACUST UNITED AC 2016; 145:459-74. [PMID: 26009541 PMCID: PMC4442791 DOI: 10.1085/jgp.201411321] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A novel category of diseases of striated muscle is proposed, the couplonopathies, as those that affect components of the couplon and thereby alter its operation. Couplons are the functional units of intracellular calcium release in excitation–contraction coupling. They comprise dihydropyridine receptors, ryanodine receptors (Ca2+ release channels), and a growing list of ancillary proteins whose alteration may lead to disease. Within a generally similar plan, the couplons of skeletal and cardiac muscle show, in a few places, marked structural divergence associated with critical differences in the mechanisms whereby they fulfill their signaling role. Most important among these are the presence of a mechanical or allosteric communication between voltage sensors and Ca2+ release channels, exclusive to the skeletal couplon, and the smaller capacity of the Ca stores in cardiac muscle, which results in greater swings of store concentration during physiological function. Consideration of these structural and functional differences affords insights into the pathogenesis of several couplonopathies. The exclusive mechanical connection of the skeletal couplon explains differences in pathogenesis between malignant hyperthermia (MH) and catecholaminergic polymorphic ventricular tachycardia (CPVT), conditions most commonly caused by mutations in homologous regions of the skeletal and cardiac Ca2+ release channels. Based on mechanistic considerations applicable to both couplons, we identify the plasmalemma as a site of secondary modifications, typically an increase in store-operated calcium entry, that are relevant in MH pathogenesis. Similar considerations help explain the different consequences that mutations in triadin and calsequestrin have in these two tissues. As more information is gathered on the composition of cardiac and skeletal couplons, this comparative and mechanistic approach to couplonopathies should be useful to understand pathogenesis, clarify diagnosis, and propose tissue-specific drug development.
Collapse
Affiliation(s)
- Eduardo Ríos
- Section of Cellular Signaling, Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612
| | - Lourdes Figueroa
- Section of Cellular Signaling, Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612
| | - Carlo Manno
- Section of Cellular Signaling, Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612
| | - Natalia Kraeva
- Malignant Hyperthermia Investigation Unit, University Health Network, Toronto General Hospital, Toronto, Ontario M5G 2C4, Canada
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, University Health Network, Toronto General Hospital, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
19
|
Hernández-Ochoa EO, Vanegas C, Iyer SR, Lovering RM, Schneider MF. Alternating bipolar field stimulation identifies muscle fibers with defective excitability but maintained local Ca(2+) signals and contraction. Skelet Muscle 2016; 6:6. [PMID: 26855765 PMCID: PMC4743112 DOI: 10.1186/s13395-016-0076-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/05/2016] [Indexed: 11/25/2022] Open
Abstract
Background Most cultured enzymatically dissociated adult myofibers exhibit spatially uniform (UNI) contractile responses and Ca2+ transients over the entire myofiber in response to electric field stimuli of either polarity applied via bipolar electrodes. However, some myofibers only exhibit contraction and Ca2+ transients at alternating (ALT) ends in response to alternating polarity field stimulation. Here, we present for the first time the methodology for identification of ALT myofibers in primary cultures and isolated muscles, as well as a study of their electrophysiological properties. Results We used high-speed confocal microscopic Ca2+ imaging, electric field stimulation, microelectrode recordings, immunostaining, and confocal microscopy to characterize the properties of action potential-induced Ca2+ transients, contractility, resting membrane potential, and staining of T-tubule voltage-gated Na+ channel distribution applied to cultured adult myofibers. Here, we show for the first time, with high temporal and spatial resolution, that normal control myofibers with UNI responses can be converted to ALT response myofibers by TTX addition or by removal of Na+ from the bathing medium, with reappearance of the UNI response on return of Na+. Our results suggest disrupted excitability as the cause of ALT behavior and indicate that the ALT response is due to local depolarization-induced Ca2+ release, whereas the UNI response is triggered by action potential propagation over the entire myofiber. Consistent with this interpretation, local depolarizing monopolar stimuli give uniform (propagated) responses in UNI myofibers, but only local responses at the electrode in ALT myofibers. The ALT responses in electrically inexcitable myofibers are consistent with expectations of current spread between bipolar stimulating electrodes, entering (hyperpolarizing) one end of a myofiber and leaving (depolarizing) the other end of the myofiber. ALT responses were also detected in some myofibers within intact isolated whole muscles from wild-type and MDX mice, demonstrating that ALT responses can be present before enzymatic dissociation. Conclusions We suggest that checking for ALT myofiber responsiveness by looking at the end of a myofiber during alternating polarity stimuli provides a test for compromised excitability of myofibers, and could be used to identify inexcitable, damaged or diseased myofibers by ALT behavior in healthy and diseased muscle. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0076-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, 108 N. Greene Street, Baltimore, MD 21201 USA
| | - Camilo Vanegas
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, 108 N. Greene Street, Baltimore, MD 21201 USA
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, 108 N. Greene Street, Baltimore, MD 21201 USA
| |
Collapse
|
20
|
Robin G, Allard B. Voltage-gated Ca(2+) influx through L-type channels contributes to sarcoplasmic reticulum Ca(2+) loading in skeletal muscle. J Physiol 2015; 593:4781-97. [PMID: 26383921 DOI: 10.1113/jp270252] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 09/08/2015] [Indexed: 12/15/2022] Open
Abstract
Muscle contraction is triggered by Ca(2+) ions released from the sarcoplasmic reticulum (SR) in response to depolarization of skeletal muscle fibres. Muscle activation is also associated with a voltage-activated trans-sarcolemmal Ca(2+) influx early identified as a current flowing through L-type Ca(2+) channels. Because removal of external Ca(2+) does not impede fibres from contracting, a negligible role was given to this voltage-activated Ca(2+) entry, although the decline of Ca(2+) release is more pronounced in the absence of Ca(2+) during long-lasting activation. Furthermore, it is not clearly established whether Ca(2+) exclusively flows through L-type channels or in addition through a parallel voltage-activated pathway distinct from L-type channels. Here, by monitoring the quenching of fura-2 fluorescence resulting from Mn(2+) influx in voltage-controlled mouse and zebrafish isolated muscle fibres, we show that the L-type current is the only contributor to Ca(2+) influx during long-lasting depolarizations in skeletal muscle. Calibration of the Mn(2+) quenching signal allowed us to estimate a mean Mn(2+) current of 0.31 ± 0.06 A F(-1) flowing through L-type channels during a train of action potentials. Measurements of SR Ca(2+) changes with fluo-5N in response to depolarization revealed that an elevated voltage-activated Ca(2+) current potentiated SR Ca(2+) loading and addition of external Mn(2+) produced quenching of fluo-5N in the SR, indicating that voltage-activated Ca(2+) /Mn(2+) influx contributes to SR Ca(2+) /Mn(2+) loading.
Collapse
Affiliation(s)
- Gaëlle Robin
- Université de Lyon, Université Lyon 1, CNRS UMR 5534, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Villeurbanne, France
| | - Bruno Allard
- Université de Lyon, Université Lyon 1, CNRS UMR 5534, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Villeurbanne, France
| |
Collapse
|
21
|
Sitsapesan R. A new look at structures and mechanisms regulating endoplasmic/sarcoplasmic reticulum Ca(2+) release in health and disease. J Physiol 2015; 593:3239-40. [PMID: 26228552 DOI: 10.1113/jphysiol.2014.281907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/03/2014] [Accepted: 10/05/2014] [Indexed: 11/08/2022] Open
Affiliation(s)
- Rebecca Sitsapesan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| |
Collapse
|
22
|
Wang L, Zhang L, Li S, Zheng Y, Yan X, Chen M, Wang H, Putney JW, Luo D. Retrograde regulation of STIM1-Orai1 interaction and store-operated Ca2+ entry by calsequestrin. Sci Rep 2015; 5:11349. [PMID: 26087026 PMCID: PMC4471903 DOI: 10.1038/srep11349] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022] Open
Abstract
Interaction between the endoplasmic reticulum (ER)-located stromal interaction molecue1 (STIM1) and the plasma membrane-located Ca2+ channel subunit, Orai1, underlies store-operated Ca2+ entry (SOCE). Calsequestrin1 (CSQ1), a sarcoplasmic reticulum Ca2+ buffering protein, inhibits SOCE, but the mechanism of action is unknown. We identified an interaction between CSQ1 and STIM1 in HEK293 cells. An increase in monomeric CSQ1 induced by depleted Ca2+ stores, or trifluoperazine (TFP), a blocker of CSQ folding and aggregation, enhanced the CSQ1-STIM1 interaction. In cells with Ca2+ stores depleted, TFP further increased CSQ1 monomerization and CSQ1-STIM1 interaction, but reduced the association of STIM1 with Orai1 and SOCE. Over-expression of CSQ1 or a C-terminal (amino acid 388–396) deletion mutant significantly promoted the association of CSQ1 with STIM1, but suppressed both STIM1-Orai1 interaction and SOCE, while over-expression of the C-terminal (amino acid 362–396) deletion mutant had no effect. The physical interaction between low polymeric forms of CSQ1 and STIM1 likely acts by interfering with STIM1 oligimerization and inhibits STIM1-Orai1 interaction, providing a brake to SOCE under physiological conditions. This novel regulatory mechanism for SOCE may also contribute to the pathological Ca2+ overload in calsequestrin deficient diseases, such as malignant hyperthermia and ventricular tachycardia.
Collapse
Affiliation(s)
- Limin Wang
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - Lane Zhang
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - Shu Li
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - Yuanyuan Zheng
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - Xinxin Yan
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - Min Chen
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - Haoyang Wang
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| | - James W Putney
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Dali Luo
- Department of Pharmacology, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
23
|
Romberg CF, Beqollari D, Meza U, Bannister RA. RGK protein-mediated impairment of slow depolarization- dependent Ca2+ entry into developing myotubes. Channels (Austin) 2015; 8:243-8. [PMID: 24476902 DOI: 10.4161/chan.27686] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Three physiological functions have been described for the skeletal muscle 1,4-dihydropyridine receptor (Ca(V)1.1):(1) voltage-sensor for excitation-contraction (EC) coupling, (2) L-type Ca(2+) channel, and (3) voltage-sensor for slow depolarization-dependent Ca(2+) entry. Members of the RGK (Rad, Rem, Rem2, Gem/Kir) family of monomeric GTP-binding proteins are potent inhibitors of the former two functions of Ca(V)1.1. However, it is not known whether the latter function that has been attributed to Ca(V)1.1 is subject to modulation by RGK proteins. Thus, the purpose of this study was to determine whether Rad, Gem and/or Rem inhibit the slowly developing, persistent Ca(2+) entry that is dependent on the voltage-sensing capability of Ca(V)1.1. As a means to investigate this question, Venus fluorescent protein-fused RGK proteins(V-Rad, V-Rem and V-Gem) were overexpressed in “normal” mouse myotubes. We observed that such overexpression of V-Rad, V-Rem or V-Gem in myotubes caused marked changes in morphology of the cells. As shown previously for YFPRem,both L-type current and EC coupling were also impaired greatly in myotubes expressing either V-Rad or V-Gem. There ductions in L-type current and EC coupling were paralleled by reductions in depolarization-induced Ca(2+) entry. Our observations provide the first evidence of modulation of this enigmatic Ca(2+) entry pathway peculiar to skeletal muscle.
Collapse
|
24
|
Characterization of excitation–contraction coupling components in human extraocular muscles. Biochem J 2015; 466:29-36. [DOI: 10.1042/bj20140970] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We show that the expression level of RyR1 in human extraocular muscles (EOMs) is low and that these muscles express different levels of proteins involved in excitation–contraction coupling (ECC) compared with leg muscles (LMs).
Collapse
|
25
|
Beqollari D, Romberg CF, Meza U, Papadopoulos S, Bannister RA. Differential effects of RGK proteins on L-type channel function in adult mouse skeletal muscle. Biophys J 2014; 106:1950-7. [PMID: 24806927 DOI: 10.1016/j.bpj.2014.03.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/24/2014] [Accepted: 03/25/2014] [Indexed: 11/19/2022] Open
Abstract
Work in heterologous systems has revealed that members of the Rad, Rem, Rem2, Gem/Kir (RGK) family of small GTP-binding proteins profoundly inhibit L-type Ca(2+) channels via three mechanisms: 1), reduction of membrane expression; 2), immobilization of the voltage-sensors; and 3), reduction of Po without impaired voltage-sensor movement. However, the question of which mode is the critical one for inhibition of L-type channels in their native environments persists. To address this conundrum in skeletal muscle, we overexpressed Rad and Rem in flexor digitorum brevis (FDB) fibers via in vivo electroporation and examined the abilities of these two RGK isoforms to modulate the L-type Ca(2+) channel (CaV1.1). We found that Rad and Rem both potently inhibit L-type current in FDB fibers. However, intramembrane charge movement was only reduced in fibers transfected with Rad; charge movement for Rem-expressing fibers was virtually identical to charge movement observed in naïve fibers. This result indicated that Rem supports inhibition solely through a mechanism that allows for translocation of CaV1.1's voltage-sensors, whereas Rad utilizes at least one mode that limits voltage-sensor movement. Because Rad and Rem differ significantly only in their amino-termini, we constructed Rad-Rem chimeras to probe the structural basis for the distinct specificities of Rad- and Rem-mediated inhibition. Using this approach, a chimera composed of the amino-terminus of Rem and the core/carboxyl-terminus of Rad inhibited L-type current without reducing charge movement. Conversely, a chimera having the amino-terminus of Rad fused to the core/carboxyl-terminus of Rem inhibited L-type current with a concurrent reduction in charge movement. Thus, we have identified the amino-termini of Rad and Rem as the structural elements dictating the specific modes of inhibition of CaV1.1.
Collapse
Affiliation(s)
- D Beqollari
- Department of Medicine-Cardiology Division, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - C F Romberg
- Department of Medicine-Cardiology Division, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - U Meza
- Department of Medicine-Cardiology Division, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado; Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - S Papadopoulos
- Institute of Vegetative Physiology, University Hospital of Cologne, Cologne, Germany
| | - R A Bannister
- Department of Medicine-Cardiology Division, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
26
|
Clark BC, Mahato NK, Nakazawa M, Law TD, Thomas JS. The power of the mind: the cortex as a critical determinant of muscle strength/weakness. J Neurophysiol 2014; 112:3219-26. [PMID: 25274345 DOI: 10.1152/jn.00386.2014] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We tested the hypothesis that the nervous system, and the cortex in particular, is a critical determinant of muscle strength/weakness and that a high level of corticospinal inhibition is an important neurophysiological factor regulating force generation. A group of healthy individuals underwent 4 wk of wrist-hand immobilization to induce weakness. Another group also underwent 4 wk of immobilization, but they also performed mental imagery of strong muscle contractions 5 days/wk. Mental imagery has been shown to activate several cortical areas that are involved with actual motor behaviors, including premotor and M1 regions. A control group, who underwent no interventions, also participated in this study. Before, immediately after, and 1 wk following immobilization, we measured wrist flexor strength, voluntary activation (VA), and the cortical silent period (SP; a measure that reflect corticospinal inhibition quantified via transcranial magnetic stimulation). Immobilization decreased strength 45.1 ± 5.0%, impaired VA 23.2 ± 5.8%, and prolonged the SP 13.5 ± 2.6%. Mental imagery training, however, attenuated the loss of strength and VA by ∼50% (23.8 ± 5.6% and 12.9 ± 3.2% reductions, respectively) and eliminated prolongation of the SP (4.8 ± 2.8% reduction). Significant associations were observed between the changes in muscle strength and VA (r = 0.56) and SP (r = -0.39). These findings suggest neurological mechanisms, most likely at the cortical level, contribute significantly to disuse-induced weakness, and that regular activation of the cortical regions via imagery attenuates weakness and VA by maintaining normal levels of inhibition.
Collapse
Affiliation(s)
- Brian C Clark
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio; Department of Biomedical Sciences, Ohio University, Athens, Ohio; Department of Geriatric Medicine, Ohio University, Athens, Ohio;
| | - Niladri K Mahato
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio
| | - Masato Nakazawa
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio; Office of Research, Ohio University, Athens, Ohio
| | - Timothy D Law
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio; Department of Family Medicine, Ohio University, Athens, Ohio; and
| | - James S Thomas
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio; Department of Biomedical Sciences, Ohio University, Athens, Ohio; School of Rehabilitation and Communication Sciences, Ohio University, Athens, Ohio
| |
Collapse
|
27
|
Identification of differentially expressed genes in breast muscle and skin fat of postnatal Pekin duck. PLoS One 2014; 9:e107574. [PMID: 25264787 PMCID: PMC4180276 DOI: 10.1371/journal.pone.0107574] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 08/20/2014] [Indexed: 11/21/2022] Open
Abstract
Lean-type Pekin duck is a commercial breed that has been obtained through long-term selection. Investigation of the differentially expressed genes in breast muscle and skin fat at different developmental stages will contribute to a comprehensive understanding of the potential mechanisms underlying the lean-type Pekin duck phenotype. In the present study, RNA-seq was performed on breast muscle and skin fat at 2-, 4- and 6-weeks of age. More than 89% of the annotated duck genes were covered by our RNA-seq dataset. Thousands of differentially expressed genes, including many important genes involved in the regulation of muscle development and fat deposition, were detected through comparison of the expression levels in the muscle and skin fat of the same time point, or the same tissue at different time points. KEGG pathway analysis showed that the differentially expressed genes clustered significantly in many muscle development and fat deposition related pathways such as MAPK signaling pathway, PPAR signaling pathway, Calcium signaling pathway, Fat digestion and absorption, and TGF-beta signaling pathway. The results presented here could provide a basis for further investigation of the mechanisms involved in muscle development and fat deposition in Pekin duck.
Collapse
|
28
|
Liu Z, Li W, Ma X, Ding N, Spallotta F, Southon E, Tessarollo L, Gaetano C, Mukouyama YS, Thiele CJ. Essential role of the zinc finger transcription factor Casz1 for mammalian cardiac morphogenesis and development. J Biol Chem 2014; 289:29801-16. [PMID: 25190801 DOI: 10.1074/jbc.m114.570416] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chromosome 1p36 deletion syndrome is one of the most common terminal deletions observed in humans and is related to congenital heart disease (CHD). However, the 1p36 genes that contribute to heart disease have not been clearly delineated. Human CASZ1 gene localizes to 1p36 and encodes a zinc finger transcription factor. Casz1 is required for Xenopus heart ventral midline progenitor cell differentiation. Whether Casz1 plays a role during mammalian heart development is unknown. Our aim is to determine 1p36 gene CASZ1 function at regulating heart development in mammals. We generated a Casz1 knock-out mouse using Casz1-trapped embryonic stem cells. Casz1 deletion in mice resulted in abnormal heart development including hypoplasia of myocardium, ventricular septal defect, and disorganized morphology. Hypoplasia of myocardium was caused by decreased cardiomyocyte proliferation. Comparative genome-wide RNA transcriptome analysis of Casz1 depleted embryonic hearts identifies abnormal expression of genes that are critical for muscular system development and function, such as muscle contraction genes TNNI2, TNNT1, and CKM; contractile fiber gene ACTA1; and cardiac arrhythmia associated ion channel coding genes ABCC9 and CACNA1D. The transcriptional regulation of some of these genes by Casz1 was also found in cellular models. Our results showed that loss of Casz1 during mouse development led to heart defect including cardiac noncompaction and ventricular septal defect, which phenocopies 1p36 deletion syndrome related CHD. This suggests that CASZ1 is a novel 1p36 CHD gene and that the abnormal expression of cardiac morphogenesis and contraction genes induced by loss of Casz1 contributes to the heart defect.
Collapse
Affiliation(s)
| | - Wenling Li
- the Laboratories of Stem Cell and Neuro-vascular Biology and
| | - Xuefei Ma
- the Molecular Cardiology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892, and
| | | | - Francesco Spallotta
- the Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany
| | - Eileen Southon
- the Mouse Cancer Genetics Program, Neural Development Section, National Cancer Institute, Bethesda, Maryland 20892
| | - Lino Tessarollo
- the Mouse Cancer Genetics Program, Neural Development Section, National Cancer Institute, Bethesda, Maryland 20892
| | - Carlo Gaetano
- the Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany
| | | | | |
Collapse
|
29
|
Calderón JC, Bolaños P, Caputo C. The excitation-contraction coupling mechanism in skeletal muscle. Biophys Rev 2014; 6:133-160. [PMID: 28509964 PMCID: PMC5425715 DOI: 10.1007/s12551-013-0135-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/06/2013] [Indexed: 12/27/2022] Open
Abstract
First coined by Alexander Sandow in 1952, the term excitation-contraction coupling (ECC) describes the rapid communication between electrical events occurring in the plasma membrane of skeletal muscle fibres and Ca2+ release from the SR, which leads to contraction. The sequence of events in twitch skeletal muscle involves: (1) initiation and propagation of an action potential along the plasma membrane, (2) spread of the potential throughout the transverse tubule system (T-tubule system), (3) dihydropyridine receptors (DHPR)-mediated detection of changes in membrane potential, (4) allosteric interaction between DHPR and sarcoplasmic reticulum (SR) ryanodine receptors (RyR), (5) release of Ca2+ from the SR and transient increase of Ca2+ concentration in the myoplasm, (6) activation of the myoplasmic Ca2+ buffering system and the contractile apparatus, followed by (7) Ca2+ disappearance from the myoplasm mediated mainly by its reuptake by the SR through the SR Ca2+ adenosine triphosphatase (SERCA), and under several conditions movement to the mitochondria and extrusion by the Na+/Ca2+ exchanger (NCX). In this text, we review the basics of ECC in skeletal muscle and the techniques used to study it. Moreover, we highlight some recent advances and point out gaps in knowledge on particular issues related to ECC such as (1) DHPR-RyR molecular interaction, (2) differences regarding fibre types, (3) its alteration during muscle fatigue, (4) the role of mitochondria and store-operated Ca2+ entry in the general ECC sequence, (5) contractile potentiators, and (6) Ca2+ sparks.
Collapse
Affiliation(s)
- Juan C Calderón
- Physiology and Biochemistry Research Group-Physis, Department of Physiology and Biochemistry, Faculty of Medicine, University of Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia.
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela.
- Departamento de Fisiología y Bioquímica, Grupo de Investigación en Fisiología y Bioquímica-Physis, Facultad de Medicina, Universidad de Antioquia, Calle 70 No 52-21, Medellín, Colombia.
| | - Pura Bolaños
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - Carlo Caputo
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| |
Collapse
|
30
|
Establishment of a human skeletal muscle-derived cell line: biochemical, cellular and electrophysiological characterization. Biochem J 2013; 455:169-77. [PMID: 23905709 DOI: 10.1042/bj20130698] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Excitation-contraction coupling is the physiological mechanism occurring in muscle cells whereby an electrical signal sensed by the dihydropyridine receptor located on the transverse tubules is transformed into a chemical gradient (Ca2+ increase) by activation of the ryanodine receptor located on the sarcoplasmic reticulum membrane. In the present study, we characterized for the first time the excitation-contraction coupling machinery of an immortalized human skeletal muscle cell line. Intracellular Ca2+ measurements showed a normal response to pharmacological activation of the ryanodine receptor, whereas 3D-SIM (super-resolution structured illumination microscopy) revealed a low level of structural organization of ryanodine receptors and dihydropyridine receptors. Interestingly, the expression levels of several transcripts of proteins involved in Ca2+ homoeostasis and differentiation indicate that the cell line has a phenotype closer to that of slow-twitch than fast-twitch muscles. These results point to the potential application of such human muscle-derived cell lines to the study of neuromuscular disorders; in addition, they may serve as a platform for the development of therapeutic strategies aimed at correcting defects in Ca2+ homoeostasis due to mutations in genes involved in Ca2+ regulation.
Collapse
|