1
|
Chen Z, Minor DL. Electrosome assembly: Structural insights from high voltage-activated calcium channel (CaV)-chaperone interactions. Biochem Soc Trans 2025; 53:BST20240422. [PMID: 39912874 DOI: 10.1042/bst20240422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025]
Abstract
Ion channels are multicomponent complexes (termed here as"electrosomes") that conduct the bioelectrical signals required for life. It has been appreciated for decades that assembly is critical for proper channel function, but knowledge of the factors that undergird this important process has been lacking. Although there are now exemplar structures of representatives of most major ion channel classes, there has been no direct structural information to inform how these complicated, multipart complexes are put together or whether they interact with chaperone proteins that aid in their assembly. Recent structural characterization of a complex of the endoplasmic membrane protein complex (EMC) chaperone and a voltage-gated calcium channel (CaV) assembly intermediate comprising the pore-forming CaVα1 and cytoplasmic CaVβ subunits offers the first structural view into the assembly of a member of the largest ion channel class, the voltagegated ion channel (VGIC) superfamily. The structure shows how the EMC remodels the CaVα1/CaVβ complex through a set of rigid body movements for handoff to the extracellular CaVα2δ subunit to complete channel assembly in a process that involves intersubunit coordination of a divalent cation and ordering of CaVα1 elements. These findings set a new framework for deciphering the structural underpinnings of ion channel biogenesis that has implications for understanding channel function, how drugs and disease mutations act, and for investigating how other membrane proteins may engage the ubiquitous EMC chaperone.
Collapse
Affiliation(s)
- Zhou Chen
- Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- Department of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- California Institute for Quantitative Biomedical Research, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- Kavli Institute for Fundamental Neuroscience, University of California-San Francisco, San Francisco, CA 94158-9001, U.S.A
- Molecular Biophysics and Integrated Bio-imaging Division Lawrence Berkeley National Laboratory, Berkeley, CA 94720 CA 94720, U.S.A
| |
Collapse
|
2
|
Martus D, Williams SK, Pichi K, Mannebach-Götz S, Kaiser N, Wardas B, Fecher-Trost C, Meyer MR, Schmitz F, Beck A, Fairless R, Diem R, Flockerzi V, Belkacemi A. Cavβ3 Contributes to the Maintenance of the Blood-Brain Barrier and Alleviates Symptoms of Experimental Autoimmune Encephalomyelitis. Arterioscler Thromb Vasc Biol 2024; 44:1833-1851. [PMID: 38957986 DOI: 10.1161/atvbaha.124.321141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tight control of cytoplasmic Ca2+ concentration in endothelial cells is essential for the regulation of endothelial barrier function. Here, we investigated the role of Cavβ3, a subunit of voltage-gated Ca2+ (Cav) channels, in modulating Ca2+ signaling in brain microvascular endothelial cells (BMECs) and how this contributes to the integrity of the blood-brain barrier. METHODS We investigated the function of Cavβ3 in BMECs by Ca2+ imaging and Western blot, examined the endothelial barrier function in vitro and the integrity of the blood-brain barrier in vivo, and evaluated disease course after induction of experimental autoimmune encephalomyelitis in mice using Cavβ3-/- (Cavβ3-deficient) mice as controls. RESULTS We identified Cavβ3 protein in BMECs, but electrophysiological recordings did not reveal significant Cav channel activity. In vivo, blood-brain barrier integrity was reduced in the absence of Cavβ3. After induction of experimental autoimmune encephalomyelitis, Cavβ3-/- mice showed earlier disease onset with exacerbated clinical disability and increased T-cell infiltration. In vitro, the transendothelial resistance of Cavβ3-/- BMEC monolayers was lower than that of wild-type BMEC monolayers, and the organization of the junctional protein ZO-1 (zona occludens-1) was impaired. Thrombin stimulates inositol 1,4,5-trisphosphate-dependent Ca2+ release, which facilitates cell contraction and enhances endothelial barrier permeability via Ca2+-dependent phosphorylation of MLC (myosin light chain). These effects were more pronounced in Cavβ3-/- than in wild-type BMECs, whereas the differences were abolished in the presence of the MLCK (MLC kinase) inhibitor ML-7. Expression of Cacnb3 cDNA in Cavβ3-/- BMECs restored the wild-type phenotype. Coimmunoprecipitation and mass spectrometry demonstrated the association of Cavβ3 with inositol 1,4,5-trisphosphate receptor proteins. CONCLUSIONS Independent of its function as a subunit of Cav channels, Cavβ3 interacts with the inositol 1,4,5-trisphosphate receptor and is involved in the tight control of cytoplasmic Ca2+ concentration and Ca2+-dependent MLC phosphorylation in BMECs, and this role of Cavβ3 in BMECs contributes to blood-brain barrier integrity and attenuates the severity of experimental autoimmune encephalomyelitis disease.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Blood-Brain Barrier/metabolism
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium Channels/genetics
- Calcium Signaling
- Capillary Permeability
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Endothelial Cells/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Myosin Light Chains/metabolism
- Myosin-Light-Chain Kinase/metabolism
- Myosin-Light-Chain Kinase/genetics
- Phosphorylation
Collapse
Affiliation(s)
- Damian Martus
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Sarah K Williams
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Kira Pichi
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Stefanie Mannebach-Götz
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Nicolas Kaiser
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Barbara Wardas
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Claudia Fecher-Trost
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Markus R Meyer
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Frank Schmitz
- Institut für Anatomie und Zellbiologie (F.S.), Universität des Saarlandes, Homburg, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Richard Fairless
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Ricarda Diem
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Anouar Belkacemi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
- Now with Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Germany (A. Belkacemi)
| |
Collapse
|
3
|
Liu G, Lv Y, Wang Y, Xu Z, Chen L, Chen S, Xie W, Feng Y, Liu J, Bai Y, He Y, Li X, Wu Q. Remote ischemic preconditioning reduces mitochondrial apoptosis mediated by calpain 1 activation in myocardial ischemia-reperfusion injury through calcium channel subunit Cacna2d3. Free Radic Biol Med 2024; 212:80-93. [PMID: 38151212 DOI: 10.1016/j.freeradbiomed.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/29/2023]
Abstract
Remote Ischemic Preconditioning (RIPC) can reduce myocardial ischemia-reperfusion injury, but its mechanism is not clear. In order to explore the mechanism of RIPC in myocardial protection, we collected myocardial specimens during cardiac surgery in children with tetralogy of Fallot for sequencing. Our study found RIPC reduces the expression of the calcium channel subunit cacna2d3, thereby impacting the function of calcium channels. As a result, calcium overload during ischemia-reperfusion is reduced, and the activation of calpain 1 is inhibited. This ultimately leads to a decrease in calpain 1 cleavage of Bax, consequently inhibiting increased mitochondrial permeability-mediated apoptosis. Notably, in both murine and human models of myocardial ischemia-reperfusion injury, RIPC inhibiting the expression of the calcium channel subunit cacna2d3 and the activation of calpain 1, improving cardiac function and histological outcomes. Overall, our findings put forth a proposed mechanism that elucidates how RIPC reduces myocardial ischemia-reperfusion injury, ultimately providing a solid theoretical foundation for the widespread clinic application of RIPC.
Collapse
Affiliation(s)
- Guoyang Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yong Lv
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yanting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Zhenzhen Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Lu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shiqiang Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Wanli Xie
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yiqi Feng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Jie Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yunxiao Bai
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yuyao He
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
4
|
Karmakar S, Basak HK, Paswan U, Saha S, Mandal SK, Chatterjee A. Design, In silico Screening, Synthesis, Characterisation and DFT-based Electronic Properties of Dihydropyridine-based Molecule as L-type Calcium Channel Blocker. Curr Comput Aided Drug Des 2024; 20:1130-1146. [PMID: 39354859 DOI: 10.2174/0115734099273719231005062524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2024]
Abstract
BACKGROUND People of all nationalities and social classes are now affected by the growing issue of hypertension. Over time, there has been a consistent rise in the fatality rate. A range of therapeutic compounds, on the other hand, are often used to handle hypertension. OBJECTIVES The objectives of this study are first to design potential antihypertensive drugs based on the DHP scaffold, secondly, to analyse drug-likeness properties of the ligands and investigate their molecular mechanisms of binding to the model protein Cav1.2 and finally to synthesise the best ligand. MATERIALS AND METHODS Due to the lack of 3D structures for human Cav1.2, the protein structure was modelled using a homology modelling approach. A protein-ligand complex's strength and binding interaction were investigated using molecular docking and molecular dynamics techniques. DFT-based electronic properties of the ligand were calculated using the M06-2X/ def2- TZVP level of theory. The SwissADME website was used to study the ADMET properties. RESULTS In this study, a series of DHP compounds (19 compounds) were properly designed to act as calcium channel blockers. Among these compounds, compound 16 showed excellent binding scores (-11.6 kcal/mol). This compound was synthesised with good yield and characterised. To assess the structural features of the synthesised molecule quantum chemical calculations were performed. CONCLUSION Based on molecular docking, molecular dynamics simulations, and drug-likeness properties of compound 16 can be used as a potential calcium channel blocker.
Collapse
Affiliation(s)
- Sujoy Karmakar
- Design, Synthesis and Simulation Laboratory, Department of Chemistry, Raiganj University, 733134, WB, India
| | - Hriday Kumar Basak
- Design, Synthesis and Simulation Laboratory, Department of Chemistry, Raiganj University, 733134, WB, India
- Department of Chemistry, Government General Degree College at Kushmandi, Dakhin Dinajpur, 733121, West Bengal, India
| | - Uttam Paswan
- Design, Synthesis and Simulation Laboratory, Department of Chemistry, Raiganj University, 733134, WB, India
| | - Soumen Saha
- Department of Chemistry, TDB College, Raniganj, Paschim Bardhaman, 713347, WB, India
| | - Samir Kumar Mandal
- Department of Chemistry, Saldiha College, Saldiha, Bankura, 722173, WB, India
| | - Abhik Chatterjee
- Design, Synthesis and Simulation Laboratory, Department of Chemistry, Raiganj University, 733134, WB, India
| |
Collapse
|
5
|
Bibollet H, Kramer A, Bannister RA, Hernández-Ochoa EO. Advances in Ca V1.1 gating: New insights into permeation and voltage-sensing mechanisms. Channels (Austin) 2023; 17:2167569. [PMID: 36642864 PMCID: PMC9851209 DOI: 10.1080/19336950.2023.2167569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/09/2023] [Indexed: 01/17/2023] Open
Abstract
The CaV1.1 voltage-gated Ca2+ channel carries L-type Ca2+ current and is the voltage-sensor for excitation-contraction (EC) coupling in skeletal muscle. Significant breakthroughs in the EC coupling field have often been close on the heels of technological advancement. In particular, CaV1.1 was the first voltage-gated Ca2+ channel to be cloned, the first ion channel to have its gating current measured and the first ion channel to have an effectively null animal model. Though these innovations have provided invaluable information regarding how CaV1.1 detects changes in membrane potential and transmits intra- and inter-molecular signals which cause opening of the channel pore and support Ca2+ release from the sarcoplasmic reticulum remain elusive. Here, we review current perspectives on this topic including the recent application of functional site-directed fluorometry.
Collapse
Affiliation(s)
- Hugo Bibollet
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Audra Kramer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Roger A. Bannister
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erick O. Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Chen Z, Mondal A, Abderemane-Ali F, Jang S, Niranjan S, Montaño JL, Zaro BW, Minor DL. EMC chaperone-Ca V structure reveals an ion channel assembly intermediate. Nature 2023; 619:410-419. [PMID: 37196677 PMCID: PMC10896479 DOI: 10.1038/s41586-023-06175-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
Voltage-gated ion channels (VGICs) comprise multiple structural units, the assembly of which is required for function1,2. Structural understanding of how VGIC subunits assemble and whether chaperone proteins are required is lacking. High-voltage-activated calcium channels (CaVs)3,4 are paradigmatic multisubunit VGICs whose function and trafficking are powerfully shaped by interactions between pore-forming CaV1 or CaV2 CaVα1 (ref. 3), and the auxiliary CaVβ5 and CaVα2δ subunits6,7. Here we present cryo-electron microscopy structures of human brain and cardiac CaV1.2 bound with CaVβ3 to a chaperone-the endoplasmic reticulum membrane protein complex (EMC)8,9-and of the assembled CaV1.2-CaVβ3-CaVα2δ-1 channel. These structures provide a view of an EMC-client complex and define EMC sites-the transmembrane (TM) and cytoplasmic (Cyto) docks; interaction between these sites and the client channel causes partial extraction of a pore subunit and splays open the CaVα2δ-interaction site. The structures identify the CaVα2δ-binding site for gabapentinoid anti-pain and anti-anxiety drugs6, show that EMC and CaVα2δ interactions with the channel are mutually exclusive, and indicate that EMC-to-CaVα2δ hand-off involves a divalent ion-dependent step and CaV1.2 element ordering. Disruption of the EMC-CaV complex compromises CaV function, suggesting that the EMC functions as a channel holdase that facilitates channel assembly. Together, the structures reveal a CaV assembly intermediate and EMC client-binding sites that could have wide-ranging implications for the biogenesis of VGICs and other membrane proteins.
Collapse
Affiliation(s)
- Zhou Chen
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Sangeeta Niranjan
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - José L Montaño
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Balyn W Zaro
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| |
Collapse
|
7
|
El Ghaleb Y, Ortner NJ, Posch W, Fernández-Quintero ML, Tuinte WE, Monteleone S, Draheim HJ, Liedl KR, Wilflingseder D, Striessnig J, Tuluc P, Flucher BE, Campiglio M. Calcium current modulation by the γ1 subunit depends on alternative splicing of CaV1.1. J Gen Physiol 2022; 154:e202113028. [PMID: 35349630 PMCID: PMC9037348 DOI: 10.1085/jgp.202113028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/08/2022] [Indexed: 01/01/2023] Open
Abstract
The skeletal muscle voltage-gated calcium channel (CaV1.1) primarily functions as a voltage sensor for excitation-contraction coupling. Conversely, its ion-conducting function is modulated by multiple mechanisms within the pore-forming α1S subunit and the auxiliary α2δ-1 and γ1 subunits. In particular, developmentally regulated alternative splicing of exon 29, which inserts 19 amino acids in the extracellular IVS3-S4 loop of CaV1.1a, greatly reduces the current density and shifts the voltage dependence of activation to positive potentials outside the physiological range. We generated new HEK293 cell lines stably expressing α2δ-1, β3, and STAC3. When the adult (CaV1.1a) and embryonic (CaV1.1e) splice variants were expressed in these cells, the difference in the voltage dependence of activation observed in muscle cells was reproduced, but not the reduced current density of CaV1.1a. Only when we further coexpressed the γ1 subunit was the current density of CaV1.1a, but not that of CaV1.1e, reduced by >50%. In addition, γ1 caused a shift of the voltage dependence of inactivation to negative voltages in both variants. Thus, the current-reducing effect of γ1, unlike its effect on inactivation, is specifically dependent on the inclusion of exon 29 in CaV1.1a. Molecular structure modeling revealed several direct ionic interactions between residues in the IVS3-S4 loop and the γ1 subunit. However, substitution of these residues by alanine, individually or in combination, did not abolish the γ1-dependent reduction of current density, suggesting that structural rearrangements in CaV1.1a induced by inclusion of exon 29 may allosterically empower the γ1 subunit to exert its inhibitory action on CaV1.1 calcium currents.
Collapse
Affiliation(s)
- Yousra El Ghaleb
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Nadine J. Ortner
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Wilfried Posch
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Wietske E. Tuinte
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Stefania Monteleone
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Henning J. Draheim
- Boehringer Ingelheim Pharma GmbH & Co KG, CNS Research, Biberach an der Riss, Germany
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Bernhard E. Flucher
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Marta Campiglio
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
8
|
Pereira da Silva EA, Martín-Aragón Baudel M, Navedo MF, Nieves-Cintrón M. Ion channel molecular complexes in vascular smooth muscle. Front Physiol 2022; 13:999369. [PMID: 36091375 PMCID: PMC9459047 DOI: 10.3389/fphys.2022.999369] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022] Open
Abstract
Ion channels that influence membrane potential and intracellular calcium concentration control vascular smooth muscle excitability. Voltage-gated calcium channels (VGCC), transient receptor potential (TRP) channels, voltage (KV), and Ca2+-activated K+ (BK) channels are key regulators of vascular smooth muscle excitability and contractility. These channels are regulated by various signaling cues, including protein kinases and phosphatases. The effects of these ubiquitous signaling molecules often depend on the formation of macromolecular complexes that provide a platform for targeting and compartmentalizing signaling events to specific substrates. This manuscript summarizes our current understanding of specific molecular complexes involving VGCC, TRP, and KV and BK channels and their contribution to regulating vascular physiology.
Collapse
|
9
|
Abstract
Each heartbeat is initiated by the action potential, an electrical signal that depolarizes the plasma membrane and activates a cycle of calcium influx via voltage-gated calcium channels, calcium release via ryanodine receptors, and calcium reuptake and efflux via calcium-ATPase pumps and sodium-calcium exchangers. Agonists of the sympathetic nervous system bind to adrenergic receptors in cardiomyocytes, which, via cascading signal transduction pathways and protein kinase A (PKA), increase the heart rate (chronotropy), the strength of myocardial contraction (inotropy), and the rate of myocardial relaxation (lusitropy). These effects correlate with increased intracellular concentration of calcium, which is required for the augmentation of cardiomyocyte contraction. Despite extensive investigations, the molecular mechanisms underlying sympathetic nervous system regulation of calcium influx in cardiomyocytes have remained elusive over the last 40 years. Recent studies have uncovered the mechanisms underlying this fundamental biologic process, namely that PKA phosphorylates a calcium channel inhibitor, Rad, thereby releasing inhibition and increasing calcium influx. Here, we describe an updated model for how signals from adrenergic agonists are transduced to stimulate calcium influx and contractility in the heart.
Collapse
Affiliation(s)
- Arianne Papa
- Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jared Kushner
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA;
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA;
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
10
|
Tuluc P, Theiner T, Jacobo-Piqueras N, Geisler SM. Role of High Voltage-Gated Ca 2+ Channel Subunits in Pancreatic β-Cell Insulin Release. From Structure to Function. Cells 2021; 10:2004. [PMID: 34440773 PMCID: PMC8393260 DOI: 10.3390/cells10082004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
The pancreatic islets of Langerhans secrete several hormones critical for glucose homeostasis. The β-cells, the major cellular component of the pancreatic islets, secrete insulin, the only hormone capable of lowering the plasma glucose concentration. The counter-regulatory hormone glucagon is secreted by the α-cells while δ-cells secrete somatostatin that via paracrine mechanisms regulates the α- and β-cell activity. These three peptide hormones are packed into secretory granules that are released through exocytosis following a local increase in intracellular Ca2+ concentration. The high voltage-gated Ca2+ channels (HVCCs) occupy a central role in pancreatic hormone release both as a source of Ca2+ required for excitation-secretion coupling as well as a scaffold for the release machinery. HVCCs are multi-protein complexes composed of the main pore-forming transmembrane α1 and the auxiliary intracellular β, extracellular α2δ, and transmembrane γ subunits. Here, we review the current understanding regarding the role of all HVCC subunits expressed in pancreatic β-cell on electrical activity, excitation-secretion coupling, and β-cell mass. The evidence we review was obtained from many seminal studies employing pharmacological approaches as well as genetically modified mouse models. The significance for diabetes in humans is discussed in the context of genetic variations in the genes encoding for the HVCC subunits.
Collapse
Affiliation(s)
- Petronel Tuluc
- Centre for Molecular Biosciences, Department of Pharmacology and Toxicology, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (T.T.); (N.J.-P.); (S.M.G.)
| | | | | | | |
Collapse
|
11
|
Sundararaj S, Ravindran A, Casarotto MG. AHNAK: The quiet giant in calcium homeostasis. Cell Calcium 2021; 96:102403. [PMID: 33813182 DOI: 10.1016/j.ceca.2021.102403] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 10/21/2022]
Abstract
The phosphoprotein AHNAK is a large, ubiquitously expressed scaffolding protein involved in mediating a host of protein-protein interactions. This enables AHNAK to participate in various multi-protein complexes thereby orchestrating a range of diverse biological processes, including tumour suppression, immune regulation and cell architecture maintenance. A less studied but nonetheless equally important function occurs in calcium homeostasis. It does so by largely interacting with the L-type voltage-gated calcium channel (LVGCC) present in the plasma membrane of excitable cells such as muscles and neurons. Several studies have characterized the underlying basis of AHNAK's functional role in calcium channel modulation, which has led to a greater understanding of this cellular process and its associated pathologies. In this article we review and examine recent advances relating to the physiological aspects of AHNAK in calcium regulation. Specifically, we will provide a broad overview of AHNAK including its structural makeup and its interaction with several isoforms of LVGCC, and how these molecular interactions regulate calcium modulation across various tissues and their implication in muscle and neuronal function.
Collapse
Affiliation(s)
- Srinivasan Sundararaj
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia.
| | - Agin Ravindran
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Marco G Casarotto
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia.
| |
Collapse
|
12
|
Papa A, Kushner J, Hennessey JA, Katchman AN, Zakharov SI, Chen BX, Yang L, Lu R, Leong S, Diaz J, Liu G, Roybal D, Liao X, del Rivero Morfin PJ, Colecraft HM, Pitt GS, Clarke O, Topkara V, Ben-Johny M, Marx SO. Adrenergic Ca V1.2 Activation via Rad Phosphorylation Converges at α 1C I-II Loop. Circ Res 2021; 128:76-88. [PMID: 33086983 PMCID: PMC7790865 DOI: 10.1161/circresaha.120.317839] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
RATIONALE Changing activity of cardiac CaV1.2 channels under basal conditions, during sympathetic activation, and in heart failure is a major determinant of cardiac physiology and pathophysiology. Although cardiac CaV1.2 channels are prominently upregulated via activation of PKA (protein kinase A), essential molecular details remained stubbornly enigmatic. OBJECTIVE The primary goal of this study was to determine how various factors converging at the CaV1.2 I-II loop interact to regulate channel activity under basal conditions, during β-adrenergic stimulation, and in heart failure. METHODS AND RESULTS We generated transgenic mice with expression of CaV1.2 α1C subunits with (1) mutations ablating interaction between α1C and β-subunits, (2) flexibility-inducing polyglycine substitutions in the I-II loop (GGG-α1C), or (3) introduction of the alternatively spliced 25-amino acid exon 9* mimicking a splice variant of α1C upregulated in the hypertrophied heart. Introducing 3 glycine residues that disrupt a rigid IS6-α-interaction domain helix markedly reduced basal open probability despite intact binding of CaVβ to α1C I-II loop and eliminated β-adrenergic agonist stimulation of CaV1.2 current. In contrast, introduction of the exon 9* splice variant in the α1C I-II loop, which is increased in ventricles of patients with end-stage heart failure, increased basal open probability but did not attenuate stimulatory response to β-adrenergic agonists when reconstituted heterologously with β2B and Rad or transgenically expressed in cardiomyocytes. CONCLUSIONS Ca2+ channel activity is dynamically modulated under basal conditions, during β-adrenergic stimulation, and in heart failure by mechanisms converging at the α1C I-II loop. CaVβ binding to α1C stabilizes an increased channel open probability gating mode by a mechanism that requires an intact rigid linker between the β-subunit binding site in the I-II loop and the channel pore. Release of Rad-mediated inhibition of Ca2+ channel activity by β-adrenergic agonists/PKA also requires this rigid linker and β-binding to α1C.
Collapse
Affiliation(s)
- Arianne Papa
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
- Department of Physiology and Cellular Biophysics
| | - Jared Kushner
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Jessica A. Hennessey
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Alexander N. Katchman
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Sergey I. Zakharov
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Bi-xing Chen
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Lin Yang
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Ree Lu
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Stephen Leong
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Johanna Diaz
- Department of Physiology and Cellular Biophysics
| | - Guoxia Liu
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Daniel Roybal
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
- Department of Pharmacology and Molecular Signaling, Columbia University, Vagelos College of Physicians and Surgeons
| | - Xianghai Liao
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | | | - Henry M. Colecraft
- Department of Physiology and Cellular Biophysics
- Department of Pharmacology and Molecular Signaling, Columbia University, Vagelos College of Physicians and Surgeons
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medical College
| | | | - Veli Topkara
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
| | | | - Steven O. Marx
- Division of Cardiology, Department of Medicine, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY 10032
- Department of Pharmacology and Molecular Signaling, Columbia University, Vagelos College of Physicians and Surgeons
| |
Collapse
|
13
|
Balderas-Villalobos J, Steele TWE, Eltit JM. Physiological and Pathological Relevance of Selective and Nonselective Ca 2+ Channels in Skeletal and Cardiac Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:225-247. [PMID: 35138617 PMCID: PMC10683374 DOI: 10.1007/978-981-16-4254-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Contraction of the striated muscle is fundamental for human existence. The action of voluntary skeletal muscle enables activities such as breathing, establishing body posture, and diverse body movements. Additionally, highly precise motion empowers communication, artistic expression, and other activities that define everyday human life. The involuntary contraction of striated muscle is the core function of the heart and is essential for blood flow. Several ion channels are important in the transduction of action potentials to cytosolic Ca2+ signals that enable muscle contraction; however, other ion channels are involved in the progression of muscle pathologies that can impair normal life or threaten it. This chapter describes types of selective and nonselective Ca2+ permeable ion channels expressed in the striated muscle, their participation in different aspects of muscle excitation and contraction, and their relevance to the progression of some pathological states.
Collapse
Affiliation(s)
- Jaime Balderas-Villalobos
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tyler W E Steele
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
14
|
Investigation of the Selectivity of L-Type Voltage-Gated Calcium Channels 1.3 for Pyrimidine-2,4,6-Triones Derivatives Based on Molecular Dynamics Simulation. Molecules 2020; 25:molecules25225440. [PMID: 33233858 PMCID: PMC7699898 DOI: 10.3390/molecules25225440] [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: 10/24/2020] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 11/17/2022] Open
Abstract
Human Cav1.3 (hCav1.3) is of great interest as a potential target for Parkinson’s disease. However, common medications like dihydropyridines (DHPs), a kind of classic calcium channel blocker, have poor selectivity to hCav1.3 in clinical treatment, mainly due to being implicated in cardiovascular side-effects mediated by human Cav1.2 (hCav1.2). Recently, pyrimidine-2,4,6-triones (PYTs) have received extensive attention as prominent selective inhibitors to hCav1.3. In this study, we describe the selectivity mechanism of PYTs for hCav1.2 and hCav1.3 based on molecular dynamic simulation methods. Our results reveal that the van der Waals (vdW) interaction was the most important force affecting selectivity. Moreover, the hydrophobic interaction was more conducive to the combination. The highly hydrophobic amino acid residues on hCav1.3, such as V162 (IR1), L303 (IR2), M481 (IR3), and F484 (IR3), provided the greatest contributions in the binding free energy. On the other hand, the substituents of a halogen-substituted aromatic ring, cycloalkyl and norbornyl on PYTs, which are pertinent to the steric hindrance of the compounds, played core roles in the selectivity and affinity for hCav1.3, whereas strong polar substituents needed to be avoided. The findings could provide valuable information for designing more effective and safe medicines for Parkinson’s disease.
Collapse
|
15
|
Wu Y, Liu H, Wang X. Cardioprotection of pharmacological postconditioning on myocardial ischemia/reperfusion injury. Life Sci 2020; 264:118628. [PMID: 33131670 DOI: 10.1016/j.lfs.2020.118628] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 12/26/2022]
Abstract
Acute myocardial infarction is associated with high rates of morbidity and mortality and can cause irreversible myocardial damage. Timely reperfusion is critical to limit infarct size and salvage the ischemic myocardium. However, reperfusion may exacerbate lethal tissue injury, a phenomenon known as myocardial ischemia/reperfusion (I/R) injury. Pharmacological postconditioning (PPC), a strategy involving medication administration before or during the early minutes of reperfusion, is more efficient and flexible than preconditioning or ischemic conditioning. Previous studies have shown that various mechanisms are involved in the effects of PPC. In this review, we summarize the relative effects and potential underlying mechanisms of PPC to provide a foundation for future research attempting to develop novel treatments against myocardial I/R injury.
Collapse
Affiliation(s)
- Yushi Wu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for cardiovascular Disease, 510282 Guangzhou, China; Sino-Japanese cooperation Platform for Translational Research in Heart Failure, 510282 Guangzhou, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
| | - Haiqiong Liu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for cardiovascular Disease, 510282 Guangzhou, China; Sino-Japanese cooperation Platform for Translational Research in Heart Failure, 510282 Guangzhou, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
| | - Xianbao Wang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, NO. 253, Gongye Avenue, 510282 Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for cardiovascular Disease, 510282 Guangzhou, China; Sino-Japanese cooperation Platform for Translational Research in Heart Failure, 510282 Guangzhou, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China.
| |
Collapse
|
16
|
Lozano Jiménez YY, Sánchez Mora RM. Canales de calcio como blanco de interés farmacológico. NOVA 2020. [DOI: 10.22490/24629448.3926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Los canales de calcio son proteínas de membrana que constituyen la vía más importante para el ingreso del ion calcio (Ca2+) a la célula. Al abrirse, permiten el ingreso selectivo del ion, iniciando una variedad de procesos como contracción muscular, secreción endocrina y liberación de neurotransmisores, entre otros. Estas proteínas se agrupan en tres categorías de acuerdo con sus propiedades estructurales y funcionales: (i) Canales de Ca2+ operados por interacción receptor-ligando (ROCC), (ii) Canales activados por parámetros físicos (Transient Receptor Potencial, TRP) y (iii) Canales de Calcio dependientes de voltaje (VDCCs), siendo estos últimos los más estudiados debido a su presencia en células excitables. Dada la importancia de Ca2+ en la fisiología celular, los canales de Ca2+ constituyen un punto de acción farmacológica importante para múltiples tratamientos y, por tanto, son objeto de estudio para el desarrollo de nuevos fármacos. El objetivo de esta revisión es explicar la importancia de los canales de Ca2+ desde una proyección farmacológica, a partir de la exploración documental de artículos publicados hasta la fecha teniendo en cuenta temas relacionados con la estructura de los canales Ca2+, sus propiedades biofísicas, localización celular, funcionamiento y su interacción farmacológica.
Collapse
|
17
|
Chambers L, Dorrance AM. Regulation of ion channels in the microcirculation by mineralocorticoid receptor activation. CURRENT TOPICS IN MEMBRANES 2020; 85:151-185. [PMID: 32402638 DOI: 10.1016/bs.ctm.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mineralocorticoid receptor (MR) has classically been studied in the renal epithelium for its role in regulating sodium and water balance and, subsequently, blood pressure. However, the MR also plays a critical role in the microvasculature by regulating ion channel expression and function. Activation of the MR by its endogenous agonist aldosterone results in translocation of the MR into the nucleus, where it can act as a transcription factor. Although most of the actions of the aldosterone can be attributed to its genomic activity though MR activation, it can also act by nongenomic mechanisms. Activation of this ubiquitous receptor increases the expression of epithelial sodium channels (ENaC) in both the endothelium and smooth muscle cells of peripheral and cerebral vessels. MR activation also regulates activity of calcium channels, calcium-activated potassium channels, and various transient receptor potential (TRP) channels. Modification of these ion channels results in a myriad of negative consequences, including impaired endothelium-dependent vasodilation, alterations in generation of myogenic tone, and increased inflammation and oxidative stress. Taken together, these studies demonstrate the importance of studying the impact of the MR on ion channel function in the vasculature. While research in this area has made advances in recent years, there are still many large gaps in knowledge that need to be filled. Crucial future directions of study include defining the molecular mechanisms involved in this interaction, as well as elucidating the potential sex differences that may exist, as these areas of understanding are currently lacking.
Collapse
Affiliation(s)
- Laura Chambers
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
18
|
Guerra MJ, González‐Jamett AM, Báez‐Matus X, Navarro‐Quezada N, Martínez AD, Neely A, Cárdenas AM. The Ca2+channel subunit CaVβ2a‐subunit down‐regulates voltage‐activated ion current densities by disrupting actin‐dependent traffic in chromaffin cells. J Neurochem 2019; 151:703-715. [DOI: 10.1111/jnc.14851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/01/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- María J. Guerra
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Arlek M. González‐Jamett
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Ximena Báez‐Matus
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Nieves Navarro‐Quezada
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Agustín D. Martínez
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Alan Neely
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Ana M. Cárdenas
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| |
Collapse
|
19
|
Takahashi K, Hayashi S, Miyajima M, Omori M, Wang J, Kaihara K, Morimatsu M, Wang C, Chen J, Iribe G, Naruse K, Sokabe M. L-type calcium channel modulates mechanosensitivity of the cardiomyocyte cell line H9c2. Cell Calcium 2019; 79:68-74. [PMID: 30836292 DOI: 10.1016/j.ceca.2019.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/01/2019] [Accepted: 02/18/2019] [Indexed: 01/08/2023]
Abstract
The application of mechanical stimuli to cells often induce increases in intracellular calcium, affecting the regulation of a variety of cell functions. Although the mechanism of mechanotransduction-induced calcium increases has not been fully resolved, the involvement of mechanosensitive ion channels in the plasma membrane and the endoplasmic reticulum has been reported. Here, we demonstrate that voltage-gated L-type calcium channels play a critical role in the mechanosensitive calcium response in H9c2 rat cardiomyocytes. The intracellular calcium level in H9c2 cells increased in a reproducible dose-dependent manner in response to uniaxial stretching. The stretch-activated calcium response (SICR) completely disappeared in calcium-free medium, whereas thapsigargin and cyclopiazonic acid, inhibitors of sarcoendoplasmic reticulum calcium ATPase, partially reduced the SICR. These findings suggest that both calcium influx across the cell membrane and calcium release from the sarcoendoplasmic reticulum are involved in the SICR. Nifedipine, diltiazem, and verapamil, inhibitors of L-type calcium channels, reduced the SICR in a dose-dependent manner. Furthermore, small interfering RNA against the L-type calcium channel α1c subunit diminished the SICR dramatically. Nifedipine also diminished the mechanosensitivity of Langendorff-perfused rat heart. These results suggest that the SICR in H9c2 cardiomyocytes involves the activation of L-type calcium channels and subsequent calcium release from the sarcoendoplasmic reticulum.
Collapse
Affiliation(s)
- Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan.
| | - Shogo Hayashi
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | - Mari Miyajima
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | - Marei Omori
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan
| | - Jing Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Cardiology, Qingdao Municipal Hospital, Qingdao, 266001, China
| | - Keiko Kaihara
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masatoshi Morimatsu
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chen Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jian Chen
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Gentaro Iribe
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Department of Physiology, Nagoya University School of Medicine, Nagoya, 466-8550, Japan; Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
20
|
Tammineni ER, Carrillo ED, Soto-Acosta R, Angel-Ambrocio AH, García MC, Bautista-Carbajal P, del Angel RM, Sánchez JA. The β
4
subunit of Ca
v
1.2 channels is required for an optimal interferon response in cardiac muscle cells. Sci Signal 2018; 11:11/560/eaaj1676. [DOI: 10.1126/scisignal.aaj1676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Eshwar R. Tammineni
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Elba D. Carrillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Rubén Soto-Acosta
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Antonio H. Angel-Ambrocio
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - María C. García
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Patricia Bautista-Carbajal
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Rosa M. del Angel
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Jorge A. Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| |
Collapse
|
21
|
Raifman TK, Kumar P, Haase H, Klussmann E, Dascal N, Weiss S. Protein kinase C enhances plasma membrane expression of cardiac L-type calcium channel, Ca V1.2. Channels (Austin) 2017; 11:604-615. [PMID: 28901828 DOI: 10.1080/19336950.2017.1369636] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
L-type-voltage-dependent Ca2+ channels (L-VDCCs; CaV1.2, α1C), crucial in cardiovascular physiology and pathology, are modulated via activation of G-protein-coupled receptors and subsequently protein kinase C (PKC). Despite extensive study, key aspects of the mechanisms leading to PKC-induced Ca2+ current increase are unresolved. A notable residue, Ser1928, located in the distal C-terminus (dCT) of α1C was shown to be phosphorylated by PKC. CaV1.2 undergoes posttranslational modifications yielding full-length and proteolytically cleaved CT-truncated forms. We have previously shown that, in Xenopus oocytes, activation of PKC enhances α1C macroscopic currents. This increase depended on the isoform of α1C expressed. Only isoforms containing the cardiac, long N-terminus (L-NT), were upregulated by PKC. Ser1928 was also crucial for the full effect of PKC. Here we report that, in Xenopus oocytes, following PKC activation the amount of α1C protein expressed in the plasma membrane (PM) increases within minutes. The increase in PM content is greater with full-length α1C than in dCT-truncated α1C, and requires Ser1928. The same was observed in HL-1 cells, a mouse atrium cell line natively expressing cardiac α1C, which undergoes the proteolytic cleavage of the dCT, thus providing a native setting for exploring the effects of PKC in cardiomyocytes. Interestingly, activation of PKC preferentially increased the PM levels of full-length, L-NT α1C. Our findings suggest that part of PKC regulation of CaV1.2 in the heart involves changes in channel's cellular fate. The mechanism of this PKC regulation appears to involve the C-terminus of α1C, possibly corroborating the previously proposed role of NT-CT interactions within α1C.
Collapse
Affiliation(s)
- Tal Keren Raifman
- a Department of Physiology and Pharmacology , Sackler School of Medicine, Tel Aviv University , Tel Aviv , Israel.,b Department of Physiotherapy , Zfat Academic College , Zfat , Israel
| | - Prabodh Kumar
- a Department of Physiology and Pharmacology , Sackler School of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Hannelore Haase
- c Max Delbruck Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Enno Klussmann
- c Max Delbruck Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Nathan Dascal
- a Department of Physiology and Pharmacology , Sackler School of Medicine, Tel Aviv University , Tel Aviv , Israel
| | - Sharon Weiss
- a Department of Physiology and Pharmacology , Sackler School of Medicine, Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
22
|
Bourdin B, Briot J, Tétreault MP, Sauvé R, Parent L. Negatively charged residues in the first extracellular loop of the L-type Ca V1.2 channel anchor the interaction with the Ca Vα2δ1 auxiliary subunit. J Biol Chem 2017; 292:17236-17249. [PMID: 28864774 PMCID: PMC5655503 DOI: 10.1074/jbc.m117.806893] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/18/2017] [Indexed: 12/20/2022] Open
Abstract
Voltage-gated L-type CaV1.2 channels in cardiomyocytes exist as heteromeric complexes. Co-expression of CaVα2δ1 with CaVβ/CaVα1 proteins reconstitutes the functional properties of native L-type currents, but the interacting domains at the CaV1.2/CaVα2δ1 interface are unknown. Here, a homology-based model of CaV1.2 identified protein interfaces between the extracellular domain of CaVα2δ1 and the extracellular loops of the CaVα1 protein in repeats I (IS1S2 and IS5S6), II (IIS5S6), and III (IIIS5S6). Insertion of a 9-residue hemagglutinin epitope in IS1S2, but not in IS5S6 or in IIS5S6, prevented the co-immunoprecipitation of CaV1.2 with CaVα2δ1. IS1S2 contains a cluster of three conserved negatively charged residues Glu-179, Asp-180, and Asp-181 that could contribute to non-bonded interactions with CaVα2δ1. Substitutions of CaV1.2 Asp-181 impaired the co-immunoprecipitation of CaVβ/CaV1.2 with CaVα2δ1 and the CaVα2δ1-dependent shift in voltage-dependent activation gating. In contrast, single substitutions in CaV1.2 in neighboring positions in the same loop (179, 180, and 182–184) did not significantly alter the functional up-regulation of CaV1.2 whole-cell currents. However, a negatively charged residue at position 180 was necessary to convey the CaVα2δ1-mediated shift in the activation gating. We also found a more modest contribution from the positively charged Arg-1119 in the extracellular pore region in repeat III of CaV1.2. We conclude that CaV1.2 Asp-181 anchors the physical interaction that facilitates the CaVα2δ1-mediated functional modulation of CaV1.2 currents. By stabilizing the first extracellular loop of CaV1.2, CaVα2δ1 may up-regulate currents by promoting conformations of the voltage sensor that are associated with the channel's open state.
Collapse
Affiliation(s)
- Benoîte Bourdin
- Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Julie Briot
- Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada.,From the Département de Pharmacologie et Physiologie, Faculté de Médecine, and
| | - Marie-Philippe Tétreault
- Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Rémy Sauvé
- From the Département de Pharmacologie et Physiologie, Faculté de Médecine, and
| | - Lucie Parent
- Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada .,From the Département de Pharmacologie et Physiologie, Faculté de Médecine, and
| |
Collapse
|
23
|
Segura E, Bourdin B, Tétreault MP, Briot J, Allen BG, Mayer G, Parent L. Proteolytic cleavage of the hydrophobic domain in the Ca Vα2δ1 subunit improves assembly and activity of cardiac Ca V1.2 channels. J Biol Chem 2017; 292:11109-11124. [PMID: 28495885 DOI: 10.1074/jbc.m117.784355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/24/2017] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated L-type CaV1.2 channels in cardiomyocytes exist as heteromeric complexes with the pore-forming CaVα1, CaVβ, and CaVα2δ1 subunits. The full complement of subunits is required to reconstitute the native-like properties of L-type Ca2+ currents, but the molecular determinants responsible for the formation of the heteromeric complex are still being studied. Enzymatic treatment with phosphatidylinositol-specific phospholipase C, a phospholipase C specific for the cleavage of glycosylphosphatidylinositol (GPI)-anchored proteins, disrupted plasma membrane localization of the cardiac CaVα2δ1 prompting us to investigate deletions of its hydrophobic transmembrane domain. Patch-clamp experiments indicated that the C-terminally cleaved CaVα2δ1 proteins up-regulate CaV1.2 channels. In contrast, deleting the residues before the single hydrophobic segment (CaVα2δ1 Δ1059-1063) impaired current up-regulation. CaVα2δ1 mutants G1060I and G1061I nearly eliminated the cell-surface fluorescence of CaVα2δ1, indicated by two-color flow cytometry assays and confocal imaging, and prevented CaVα2δ1-mediated increase in peak current density and modulation of the voltage-dependent gating of CaV1.2. These impacts were specific to substitutions with isoleucine residues because functional modulation was partially preserved in CaVα2δ1 G1060A and G1061A proteins. Moreover, C-terminal fragments exhibited significantly altered mobility in denatured immunoblots of CaVα2δ1 G1060I and CaVα2δ1 G1061I, suggesting that these mutant proteins were impaired in proteolytic processing. Finally, CaVα2δ1 Δ1059-1063, but not CaVα2δ1 G1060A, failed to co-immunoprecipitate with CaV1.2. Altogether, our data support a model in which small neutral hydrophobic residues facilitate the post-translational cleavage of the CaVα2δ1 subunit at the predicted membrane interface and further suggest that preventing GPI anchoring of CaVα2δ1 averts its cell-surface expression, its interaction with CaVα1, and modulation of CaV1.2 currents.
Collapse
Affiliation(s)
- Emilie Segura
- From the Départements de Pharmacologie et Physiologie and.,the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Benoîte Bourdin
- the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Marie-Philippe Tétreault
- the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Julie Briot
- From the Départements de Pharmacologie et Physiologie and.,the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Bruce G Allen
- the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada.,Médecine, Faculté de Médecine
| | - Gaétan Mayer
- the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada.,the Faculté de Pharmacie, and
| | - Lucie Parent
- From the Départements de Pharmacologie et Physiologie and .,the Centre de Recherche de l'Institut de Cardiologie de Montréal, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
24
|
Wang Y, Fehlhaber KE, Sarria I, Cao Y, Ingram NT, Guerrero-Given D, Throesch B, Baldwin K, Kamasawa N, Ohtsuka T, Sampath AP, Martemyanov KA. The Auxiliary Calcium Channel Subunit α2δ4 Is Required for Axonal Elaboration, Synaptic Transmission, and Wiring of Rod Photoreceptors. Neuron 2017; 93:1359-1374.e6. [PMID: 28262416 PMCID: PMC5364038 DOI: 10.1016/j.neuron.2017.02.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 12/31/2016] [Accepted: 02/08/2017] [Indexed: 11/24/2022]
Abstract
Neural circuit wiring relies on selective synapse formation whereby a presynaptic release apparatus is matched with its cognate postsynaptic machinery. At metabotropic synapses, the molecular mechanisms underlying this process are poorly understood. In the mammalian retina, rod photoreceptors form selective contacts with rod ON-bipolar cells by aligning the presynaptic voltage-gated Ca2+ channel directing glutamate release (CaV1.4) with postsynaptic mGluR6 receptors. We show this coordination requires an extracellular protein, α2δ4, which complexes with CaV1.4 and the rod synaptogenic mediator, ELFN1, for trans-synaptic alignment with mGluR6. Eliminating α2δ4 in mice abolishes rod synaptogenesis and synaptic transmission to rod ON-bipolar cells, and disrupts postsynaptic mGluR6 clustering. We further find that in rods, α2δ4 is crucial for organizing synaptic ribbons and setting CaV1.4 voltage sensitivity. In cones, α2δ4 is essential for CaV1.4 function, but is not required for ribbon organization, synaptogenesis, or synaptic transmission. These findings offer insights into retinal pathologies associated with α2δ4 dysfunction.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Katherine E Fehlhaber
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ignacio Sarria
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Norianne T Ingram
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Debbie Guerrero-Given
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Ben Throesch
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92121, USA
| | - Kristin Baldwin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92121, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
25
|
Chi CH, Tang CY, Pan CY. Calmodulin modulates the Ca 2+-dependent inactivation and expression level of bovine Ca V2.2 expressed in HEK293T cells. IBRO Rep 2017; 2:63-71. [PMID: 30135934 PMCID: PMC6084911 DOI: 10.1016/j.ibror.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/27/2017] [Accepted: 03/10/2017] [Indexed: 01/31/2023] Open
Abstract
Ca2+ influx through voltage-gated Ca2+ channels (CaVs) at the plasma membrane is the major pathway responsible for the elevation of the intracellular Ca2+ concentration ([Ca2+]i), which activates various physiological activities. Calmodulin (CaM) is known to be involved in the Ca2+-dependent inactivation (CDI) of several types of CaVs; however, little is known about how CaM modulates CaV2.2. Here, we expressed CaV2.2 with CaM or CaM mutants with a Ca2+-binding deficiency in HEK293T cells and measured the currents to characterize the CDI. The results showed that CaV2.2 displayed a fast inactivation with Ca2+ but not Ba2+ as the charge carrier; when CaV2.2 was co-expressed with CaM mutants with a Ca2+-binding deficiency, the level of inactivation decreased. Using glutathione S-transferase-tagged CaM or CaM mutants as the bait, we found that CaM could interact with the intracellular C-terminal fragment of CaV2.2 in the presence or absence of Ca2+. However, CaM and its mutants could not interact with this fragment when mutations were generated in the conserved amino acid residues of the CaM-binding site. CaV2.2 with mutations in the CaM-binding site showed a greatly reduced current that could be rescued by CaM12 (Ca2+-binding deficiency at the N-lobe) overexpression; in addition, CaM12 enhanced the total expression level of CaV2.2, but the ratio of CaV2.2 present in the membrane to the total fraction remained unchanged. Together, our data suggest that CaM, with different Ca2+-binding abilities, modulates not only the inactivation of CaV2.2 but also its expression to regulate Ca2+-related physiological activities.
Collapse
Affiliation(s)
- Chih-Hung Chi
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
| | - Chien-Yuan Pan
- Department of Life Science, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
26
|
Cserne Szappanos H, Viola H, Hool LC. L-type calcium channel: Clarifying the "oxygen sensing hypothesis". Int J Biochem Cell Biol 2017; 86:32-36. [PMID: 28323207 DOI: 10.1016/j.biocel.2017.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 11/30/2022]
Abstract
The heart is able to respond acutely to changes in oxygen tension. Since ion channels can respond rapidly to stimuli, the "ion channel oxygen sensing hypothesis" has been proposed to explain acute adaptation of cells to changes in oxygen demand. However the exact mechanism for oxygen sensing continues to be debated. Mitochondria consume the lion's share of oxygen in the heart, fuelling the production of ATP that drives excitation and contraction. Mitochondria also produce reactive oxygen species that are capable of altering the redox state of proteins. The cardiac L-type calcium channel is responsible for maintaining excitation and contraction. Recently, the reactive cysteine on the cardiac L-type calcium channel was identified. These data clarified that the channel does not respond directly to changes in oxygen tension, but rather responds to cellular redox state. This leads to acute alterations in cell signalling responsible for the development of arrhythmias and pathology.
Collapse
Affiliation(s)
- Henrietta Cserne Szappanos
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, WA, Australia
| | - Helena Viola
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, WA, Australia
| | - Livia C Hool
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, WA, Australia; Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
27
|
Trafficking of neuronal calcium channels. Neuronal Signal 2017; 1:NS20160003. [PMID: 32714572 PMCID: PMC7373241 DOI: 10.1042/ns20160003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 01/20/2017] [Accepted: 01/19/2017] [Indexed: 12/18/2022] Open
Abstract
Neuronal voltage-gated calcium channels (VGCCs) serve complex yet essential physiological functions via their pivotal role in translating electrical signals into intracellular calcium elevations and associated downstream signalling pathways. There are a number of regulatory mechanisms to ensure a dynamic control of the number of channels embedded in the plasma membrane, whereas alteration of the surface expression of VGCCs has been linked to various disease conditions. Here, we provide an overview of the mechanisms that control the trafficking of VGCCs to and from the plasma membrane, and discuss their implication in pathophysiological conditions and their potential as therapeutic targets.
Collapse
|
28
|
Rusconi F, Ceriotti P, Miragoli M, Carullo P, Salvarani N, Rocchetti M, Di Pasquale E, Rossi S, Tessari M, Caprari S, Cazade M, Kunderfranco P, Chemin J, Bang ML, Polticelli F, Zaza A, Faggian G, Condorelli G, Catalucci D. Peptidomimetic Targeting of Cavβ2 Overcomes Dysregulation of the L-Type Calcium Channel Density and Recovers Cardiac Function. Circulation 2016; 134:534-46. [PMID: 27486162 DOI: 10.1161/circulationaha.116.021347] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND L-type calcium channels (LTCCs) play important roles in regulating cardiomyocyte physiology, which is governed by appropriate LTCC trafficking to and density at the cell surface. Factors influencing the expression, half-life, subcellular trafficking, and gating of LTCCs are therefore critically involved in conditions of cardiac physiology and disease. METHODS Yeast 2-hybrid screenings, biochemical and molecular evaluations, protein interaction assays, fluorescence microscopy, structural molecular modeling, and functional studies were used to investigate the molecular mechanisms through which the LTCC Cavβ2 chaperone regulates channel density at the plasma membrane. RESULTS On the basis of our previous results, we found a direct linear correlation between the total amount of the LTCC pore-forming Cavα1.2 and the Akt-dependent phosphorylation status of Cavβ2 both in a mouse model of diabetic cardiac disease and in 6 diabetic and 7 nondiabetic cardiomyopathy patients with aortic stenosis undergoing aortic valve replacement. Mechanistically, we demonstrate that a conformational change in Cavβ2 triggered by Akt phosphorylation increases LTCC density at the cardiac plasma membrane, and thus the inward calcium current, through a complex pathway involving reduction of Cavα1.2 retrograde trafficking and protein degradation through the prevention of dynamin-mediated LTCC endocytosis; promotion of Cavα1.2 anterograde trafficking by blocking Kir/Gem-dependent sequestration of Cavβ2, thus facilitating the chaperoning of Cavα1.2; and promotion of Cavα1.2 transcription by the prevention of Kir/Gem-mediated shuttling of Cavβ2 to the nucleus, where it limits the transcription of Cavα1.2 through recruitment of the heterochromatin protein 1γ epigenetic repressor to the Cacna1c promoter. On the basis of this mechanism, we developed a novel mimetic peptide that, through targeting of Cavβ2, corrects LTCC life-cycle alterations, facilitating the proper function of cardiac cells. Delivery of mimetic peptide into a mouse model of diabetic cardiac disease associated with LTCC abnormalities restored impaired calcium balance and recovered cardiac function. CONCLUSIONS We have uncovered novel mechanisms modulating LTCC trafficking and life cycle and provide proof of concept for the use of Cavβ2 mimetic peptide as a novel therapeutic tool for the improvement of cardiac conditions correlated with alterations in LTCC levels and function.
Collapse
Affiliation(s)
- Francesca Rusconi
- From Humanitas Clinical and Research Center, Rozzano, Milan, Italy (F.R., P. Ceriotti, M.M., P. Carullo, N.S., E.D.P., P.K., M.-L.B., G.C., D.C.); Institute of Genetic and Biomedical Research UOS Milan National Research Council, Milan, Italy (F.R., P. Carullo, N.S., E.D.P., M.-L.B., D.C.); Department of Biotechnologies and Biosciences, University of Milan-Bicocca, Milan, Italy (M.R., A.Z.); Departments of Life Sciences (S.R.) and Clinical and Experimental Medicine (M.M.), University of Parma, Parma, Italy; University Hospital of Verona, Division of Cardiac Surgery, Verona, Italy (M.T., G.F.); Department of Sciences, University of Roma Tre, Rome, Italy (S.C., F.P.); University of Montpellier, CNRS UMR 5203, INSERM, Department of Neuroscience, Institute for Functional Genomics, LabEx Ion Channel Science and Therapeutics, Montpellier, France (M.C., J.C.); and National Institute of Nuclear Physics, Rome Tre Section, Rome, Italy (F.P.)
| | - Paola Ceriotti
- From Humanitas Clinical and Research Center, Rozzano, Milan, Italy (F.R., P. Ceriotti, M.M., P. Carullo, N.S., E.D.P., P.K., M.-L.B., G.C., D.C.); Institute of Genetic and Biomedical Research UOS Milan National Research Council, Milan, Italy (F.R., P. Carullo, N.S., E.D.P., M.-L.B., D.C.); Department of Biotechnologies and Biosciences, University of Milan-Bicocca, Milan, Italy (M.R., A.Z.); Departments of Life Sciences (S.R.) and Clinical and Experimental Medicine (M.M.), University of Parma, Parma, Italy; University Hospital of Verona, Division of Cardiac Surgery, Verona, Italy (M.T., G.F.); Department of Sciences, University of Roma Tre, Rome, Italy (S.C., F.P.); University of Montpellier, CNRS UMR 5203, INSERM, Department of Neuroscience, Institute for Functional Genomics, LabEx Ion Channel Science and Therapeutics, Montpellier, France (M.C., J.C.); and National Institute of Nuclear Physics, Rome Tre Section, Rome, Italy (F.P.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Dolphin AC. Voltage-gated calcium channels and their auxiliary subunits: physiology and pathophysiology and pharmacology. J Physiol 2016; 594:5369-90. [PMID: 27273705 PMCID: PMC5043047 DOI: 10.1113/jp272262] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Voltage‐gated calcium channels are essential players in many physiological processes in excitable cells. There are three main subdivisions of calcium channel, defined by the pore‐forming α1 subunit, the CaV1, CaV2 and CaV3 channels. For all the subtypes of voltage‐gated calcium channel, their gating properties are key for the precise control of neurotransmitter release, muscle contraction and cell excitability, among many other processes. For the CaV1 and CaV2 channels, their ability to reach their required destinations in the cell membrane, their activation and the fine tuning of their biophysical properties are all dramatically influenced by the auxiliary subunits that associate with them. Furthermore, there are many diseases, both genetic and acquired, involving voltage‐gated calcium channels. This review will provide a general introduction and then concentrate particularly on the role of auxiliary α2δ subunits in both physiological and pathological processes involving calcium channels, and as a therapeutic target.
![]()
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
30
|
Markandeya YS, Kamp TJ. Rational strategy to stop arrhythmias: Early afterdepolarizations and L-type Ca2+ current. ACTA ACUST UNITED AC 2016; 145:475-9. [PMID: 26009542 PMCID: PMC4442792 DOI: 10.1085/jgp.201511429] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yogananda S Markandeya
- Department of Medicine and Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705 Department of Medicine and Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Timothy J Kamp
- Department of Medicine and Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705 Department of Medicine and Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
31
|
Tétreault MP, Bourdin B, Briot J, Segura E, Lesage S, Fiset C, Parent L. Identification of Glycosylation Sites Essential for Surface Expression of the CaVα2δ1 Subunit and Modulation of the Cardiac CaV1.2 Channel Activity. J Biol Chem 2016; 291:4826-43. [PMID: 26742847 DOI: 10.1074/jbc.m115.692178] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Indexed: 12/15/2022] Open
Abstract
Alteration in the L-type current density is one aspect of the electrical remodeling observed in patients suffering from cardiac arrhythmias. Changes in channel function could result from variations in the protein biogenesis, stability, post-translational modification, and/or trafficking in any of the regulatory subunits forming cardiac L-type Ca(2+) channel complexes. CaVα2δ1 is potentially the most heavily N-glycosylated subunit in the cardiac L-type CaV1.2 channel complex. Here, we show that enzymatic removal of N-glycans produced a 50-kDa shift in the mobility of cardiac and recombinant CaVα2δ1 proteins. This change was also observed upon simultaneous mutation of the 16 Asn sites. Nonetheless, the mutation of only 6/16 sites was sufficient to significantly 1) reduce the steady-state cell surface fluorescence of CaVα2δ1 as characterized by two-color flow cytometry assays and confocal imaging; 2) decrease protein stability estimated from cycloheximide chase assays; and 3) prevent the CaVα2δ1-mediated increase in the peak current density and voltage-dependent gating of CaV1.2. Reversing the N348Q and N812Q mutations in the non-operational sextuplet Asn mutant protein partially restored CaVα2δ1 function. Single mutation N663Q and double mutations N348Q/N468Q, N348Q/N812Q, and N468Q/N812Q decreased protein stability/synthesis and nearly abolished steady-state cell surface density of CaVα2δ1 as well as the CaVα2δ1-induced up-regulation of L-type currents. These results demonstrate that Asn-663 and to a lesser extent Asn-348, Asn-468, and Asn-812 contribute to protein stability/synthesis of CaVα2δ1, and furthermore that N-glycosylation of CaVα2δ1 is essential to produce functional L-type Ca(2+) channels.
Collapse
Affiliation(s)
| | - Benoîte Bourdin
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Julie Briot
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Emilie Segura
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Sylvie Lesage
- Départment de Microbiologie, Infectiologie, and Immunologie, Faculté de Médecine, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Céline Fiset
- Faculté de Pharmacie, Institut de Cardiologie de Montréal and
| | - Lucie Parent
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| |
Collapse
|
32
|
Jha A, Singh AK, Weissgerber P, Freichel M, Flockerzi V, Flavell RA, Jha MK. Essential roles for Cavβ2 and Cav1 channels in thymocyte development and T cell homeostasis. Sci Signal 2015; 8:ra103. [DOI: 10.1126/scisignal.aac7538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
33
|
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280.
| |
Collapse
|
34
|
Voltage-gated calcium channels: Determinants of channel function and modulation by inorganic cations. Prog Neurobiol 2015; 129:1-36. [PMID: 25817891 DOI: 10.1016/j.pneurobio.2014.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 12/15/2014] [Accepted: 12/27/2014] [Indexed: 11/20/2022]
Abstract
Voltage-gated calcium channels (VGCCs) represent a key link between electrical signals and non-electrical processes, such as contraction, secretion and transcription. Evolved to achieve high rates of Ca(2+)-selective flux, they possess an elaborate mechanism for selection of Ca(2+) over foreign ions. It has been convincingly linked to competitive binding in the pore, but the fundamental question of how this is reconcilable with high rates of Ca(2+) transfer remains unanswered. By virtue of their similarity to Ca(2+), polyvalent cations can interfere with the function of VGCCs and have proven instrumental in probing the mechanisms underlying selective permeation. Recent emergence of crystallographic data on a set of Ca(2+)-selective model channels provides a structural framework for permeation in VGCCs, and warrants a reconsideration of their diverse modulation by polyvalent cations, which can be roughly separated into three general mechanisms: (I) long-range interactions with charged regions on the surface, affecting the local potential sensed by the channel or influencing voltage-sensor movement by repulsive forces (electrostatic effects), (II) short-range interactions with sites in the ion-conducting pathway, leading to physical obstruction of the channel (pore block), and in some cases (III) short-range interactions with extracellular binding sites, leading to non-electrostatic modifications of channel gating (allosteric effects). These effects, together with the underlying molecular modifications, provide valuable insights into the function of VGCCs, and have important physiological and pathophysiological implications. Allosteric suppression of some of the pore-forming Cavα1-subunits (Cav2.3, Cav3.2) by Zn(2+) and Cu(2+) may play a major role for the regulation of excitability by endogenous transition metal ions. The fact that these ions can often traverse VGCCs can contribute to the detrimental intracellular accumulation of metal ions following excessive release of endogenous Cu(2+) and Zn(2+) or exposure to non-physiological toxic metal ions.
Collapse
|
35
|
Olson CR, Hodges LK, Mello CV. Dynamic gene expression in the song system of zebra finches during the song learning period. Dev Neurobiol 2015; 75:1315-38. [PMID: 25787707 DOI: 10.1002/dneu.22286] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
The brain circuitry that controls song learning and production undergoes marked changes in morphology and connectivity during the song learning period in juvenile zebra finches, in parallel to the acquisition, practice and refinement of song. Yet, the genetic programs and timing of regulatory change that establish the neuronal connectivity and plasticity during this critical learning period remain largely undetermined. To address this question, we used in situ hybridization to compare the expression patterns of a set of 30 known robust molecular markers of HVC and/or area X, major telencephalic song nuclei, between adult and juvenile male zebra finches at different ages during development (20, 35, 50 days post-hatch, dph). We found that several of the genes examined undergo substantial changes in expression within HVC or its surrounds, and/or in other song nuclei. They fit into broad patterns of regulation, including those whose expression within HVC during this period increases (COL12A1, COL 21A1, MPZL1, PVALB, and CXCR7) or decreases (e.g., KCNT2, SAP30L), as well as some that show decreased expression in the surrounding tissue with little change within song nuclei (e.g. SV2B, TAC1). These results reveal a broad range of molecular changes that occur in the song system in concert with the song learning period. Some of the genes and pathways identified are potential modulators of the developmental changes associated with the emergence of the adult properties of the song control system, and/or the acquisition of learned vocalizations in songbirds.
Collapse
Affiliation(s)
- Christopher R Olson
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, Oregon, 97239-3098
| | - Lisa K Hodges
- Biology Department, Lewis and Clark College, 0615 S.W. Palatine Hill Road, Portland, Oregon 97219
| | - Claudio V Mello
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, Oregon, 97239-3098
| |
Collapse
|
36
|
Balycheva M, Faggian G, Glukhov AV, Gorelik J. Microdomain-specific localization of functional ion channels in cardiomyocytes: an emerging concept of local regulation and remodelling. Biophys Rev 2015; 7:43-62. [PMID: 28509981 PMCID: PMC5425752 DOI: 10.1007/s12551-014-0159-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/18/2014] [Indexed: 12/26/2022] Open
Abstract
Cardiac excitation involves the generation of action potential by individual cells and the subsequent conduction of the action potential from cell to cell through intercellular gap junctions. Excitation of the cellular membrane results in opening of the voltage-gated L-type calcium ion (Ca2+) channels, thereby allowing a small amount of Ca2+ to enter the cell, which in turn triggers the release of a much greater amount of Ca2+ from the sarcoplasmic reticulum, the intracellular Ca2+ store, and gives rise to the systolic Ca2+ transient and contraction. These processes are highly regulated by the autonomic nervous system, which ensures the acute and reliable contractile function of the heart and the short-term modulation of this function upon changes in heart rate or workload. It has recently become evident that discrete clusters of different ion channels and regulatory receptors are present in the sarcolemma, where they form an interacting network and work together as a part of a macro-molecular signalling complex which in turn allows the specificity, reliability and accuracy of the autonomic modulation of the excitation-contraction processes by a variety of neurohormonal pathways. Disruption in subcellular targeting of ion channels and associated signalling proteins may contribute to the pathophysiology of a variety of cardiac diseases, including heart failure and certain arrhythmias. Recent methodological advances have made it possible to routinely image the topography of live cardiomyocytes, allowing the study of clustering functional ion channels and receptors as well as their coupling within a specific microdomain. In this review we highlight the emerging understanding of the functionality of distinct subcellular microdomains in cardiac myocytes (e.g. T-tubules, lipid rafts/caveolae, costameres and intercalated discs) and their functional role in the accumulation and regulation of different subcellular populations of sodium, Ca2+ and potassium ion channels and their contributions to cellular signalling and cardiac pathology.
Collapse
Affiliation(s)
- Marina Balycheva
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
- Cardiosurgery Department, University of Verona School of Medicine, Verona, Italy
| | - Giuseppe Faggian
- Cardiosurgery Department, University of Verona School of Medicine, Verona, Italy
| | - Alexey V Glukhov
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| | - Julia Gorelik
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, 4th Floor National Heart and Lung Institute, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
37
|
Bourdin B, Shakeri B, Tétreault MP, Sauvé R, Lesage S, Parent L. Functional characterization of CaVα2δ mutations associated with sudden cardiac death. J Biol Chem 2014; 290:2854-69. [PMID: 25527503 DOI: 10.1074/jbc.m114.597930] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
L-type Ca(2+) channels play a critical role in cardiac rhythmicity. These ion channels are oligomeric complexes formed by the pore-forming CaVα1 with the auxiliary CaVβ and CaVα2δ subunits. CaVα2δ increases the peak current density and improves the voltage-dependent activation gating of CaV1.2 channels without increasing the surface expression of the CaVα1 subunit. The functional impact of genetic variants of CACNA2D1 (the gene encoding for CaVα2δ), associated with shorter repolarization QT intervals (the time interval between the Q and the T waves on the cardiac electrocardiogram), was investigated after recombinant expression of the full complement of L-type CaV1.2 subunits in human embryonic kidney 293 cells. By performing side-by-side high resolution flow cytometry assays and whole-cell patch clamp recordings, we revealed that the surface density of the CaVα2δ wild-type protein correlates with the peak current density. Furthermore, the cell surface density of CaVα2δ mutants S755T, Q917H, and S956T was not significantly different from the cell surface density of the CaVα2δ wild-type protein expressed under the same conditions. In contrast, the cell surface expression of CaVα2δ D550Y, CaVα2δ S709N, and the double mutant D550Y/Q917H was reduced, respectively, by ≈30-33% for the single mutants and by 60% for the latter. The cell surface density of D550Y/Q917H was more significantly impaired than protein stability, suggesting that surface trafficking of CaVα2δ was disrupted by the double mutation. Co-expression with D550Y/Q917H significantly decreased CaV1.2 currents as compared with results obtained with CaVα2δ wild type. It is concluded that D550Y/Q917H reduced inward Ca(2+) currents through a defect in the cell surface trafficking of CaVα2δ. Altogether, our results provide novel insight in the molecular mechanism underlying the modulation of CaV1.2 currents by CaVα2δ.
Collapse
Affiliation(s)
- Benoîte Bourdin
- From the Département de Physiologie, Montreal Heart Institute Research Centre, and
| | - Behzad Shakeri
- From the Département de Physiologie, Montreal Heart Institute Research Centre, and
| | | | - Rémy Sauvé
- From the Département de Physiologie, Montreal Heart Institute Research Centre, and
| | - Sylvie Lesage
- Département de Microbiologie, Infectiologie et Immunologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Lucie Parent
- From the Département de Physiologie, Montreal Heart Institute Research Centre, and
| |
Collapse
|
38
|
Ruan J, Liu X, Xiong X, Zhang C, Li J, Zheng H, Huang C, Shi Q, Weng Y. miR‑107 promotes the erythroid differentiation of leukemia cells via the downregulation of Cacna2d1. Mol Med Rep 2014; 11:1334-9. [PMID: 25373460 DOI: 10.3892/mmr.2014.2865] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 09/12/2014] [Indexed: 11/06/2022] Open
Abstract
microRNAs (miRNAs) have been reported to be involved in various human diseases. They may have uses in diagnosis and as therapeutic targets, thus the discovery of novel miRNAs has the potential to provide clinical tools or shed light on novel mechanisms. In the current study, miR‑107 was revealed to be downregulated in chronic myeloid leukemia cells. Overexpression of miR‑107 in K562 and KCL‑22 chronic myeloid leukemia cells promotes erythroid differentiation, while having no effect on cell proliferation. Further bioinformatics predicted that one target of miR‑107 may be Cacna2d1, a calcium channel protein. A luciferase reporter assay and quantitative polymerase chain reaction were utilized to confirm that Cacna2d1 is a target molecule of miR-107. The effect of miR‑107 on K562 and KCL‑22 cells was mediated through the downregulation of Cacna2d1, as rescued expression of Cacna2d1 reversed the effects of miR‑107. In summary, the current study identified a novel miRNA that is involved in chronic myeloid leukemia cell erythroid differentiation and the associated mechanisms, making it a potential therapeutic target in the treatment of chronic myeloid leukemia.
Collapse
Affiliation(s)
- Jie Ruan
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xinguang Liu
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xingdong Xiong
- Institute of Aging Research, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Chunlong Zhang
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jiangbin Li
- Institute of Aging Research, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Huiling Zheng
- Institute of Aging Research, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Chirong Huang
- Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Qiong Shi
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yaguang Weng
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
39
|
Mutational Consequences of Aberrant Ion Channels in Neurological Disorders. J Membr Biol 2014; 247:1083-127. [DOI: 10.1007/s00232-014-9716-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 07/25/2014] [Indexed: 12/25/2022]
|
40
|
Viola HM, Jordan MC, Roos KP, Hool LC. Decreased myocardial injury and improved contractility after administration of a peptide derived against the alpha-interacting domain of the L-type calcium channel. J Am Heart Assoc 2014; 3:e000961. [PMID: 24958783 PMCID: PMC4309103 DOI: 10.1161/jaha.114.000961] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Myocardial infarction remains the leading cause of morbidity and mortality associated with coronary artery disease. The L‐type calcium channel (ICa‐L) is critical to excitation and contraction. Activation of the channel also alters mitochondrial function. Here, we investigated whether application of a alpha‐interacting domain/transactivator of transcription (AID‐TAT) peptide, which immobilizes the auxiliary β2 subunit of the channel and decreases metabolic demand, could alter mitochondrial function and myocardial injury. Methods and Results Treatment with AID‐TAT peptide decreased ischemia‐reperfusion injury in guinea‐pig hearts ex vivo (n=11) and in rats in vivo (n=9) assessed with uptake of nitroblue tetrazolium, release of creatine kinase, and lactate dehydrogenase. Contractility (assessed with catheterization of the left ventricle) was improved after application of AID‐TAT peptide in hearts ex vivo (n=6) and in vivo (n=8) up to 12 weeks before sacrifice. In search of the mechanism for the effect, we found that intracellular calcium ([Ca2+]i, Fura‐2), superoxide production (dihydroethidium fluorescence), mitochondrial membrane potential (Ψm, JC‐1 fluorescence), reduced nicotinamide adenine dinucleotide production, and flavoprotein oxidation (autofluorescence) are decreased after application of AID‐TAT peptide. Conclusions Application of AID‐TAT peptide significantly decreased infarct size and supported contractility up to 12 weeks postcoronary artery occlusion as a result of a decrease in metabolic demand during reperfusion.
Collapse
Affiliation(s)
- Helena M Viola
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia (H.M.V., L.C.H.)
| | - Maria C Jordan
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.C.J., K.P.R.)
| | - Kenneth P Roos
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.C.J., K.P.R.)
| | - Livia C Hool
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, WA, Australia (H.M.V., L.C.H.)
| |
Collapse
|
41
|
Neely A, Hidalgo P. Structure-function of proteins interacting with the α1 pore-forming subunit of high-voltage-activated calcium channels. Front Physiol 2014; 5:209. [PMID: 24917826 PMCID: PMC4042065 DOI: 10.3389/fphys.2014.00209] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/15/2014] [Indexed: 11/13/2022] Open
Abstract
Openings of high-voltage-activated (HVA) calcium channels lead to a transient increase in calcium concentration that in turn activate a plethora of cellular functions, including muscle contraction, secretion and gene transcription. To coordinate all these responses calcium channels form supramolecular assemblies containing effectors and regulatory proteins that couple calcium influx to the downstream signal cascades and to feedback elements. According to the original biochemical characterization of skeletal muscle Dihydropyridine receptors, HVA calcium channels are multi-subunit protein complexes consisting of a pore-forming subunit (α1) associated with four additional polypeptide chains β, α2, δ, and γ, often referred to as accessory subunits. Twenty-five years after the first purification of a high-voltage calcium channel, the concept of a flexible stoichiometry to expand the repertoire of mechanisms that regulate calcium channel influx has emerged. Several other proteins have been identified that associate directly with the α1-subunit, including calmodulin and multiple members of the small and large GTPase family. Some of these proteins only interact with a subset of α1-subunits and during specific stages of biogenesis. More strikingly, most of the α1-subunit interacting proteins, such as the β-subunit and small GTPases, regulate both gating and trafficking through a variety of mechanisms. Modulation of channel activity covers almost all biophysical properties of the channel. Likewise, regulation of the number of channels in the plasma membrane is performed by altering the release of the α1-subunit from the endoplasmic reticulum, by reducing its degradation or enhancing its recycling back to the cell surface. In this review, we discuss the structural basis, interplay and functional role of selected proteins that interact with the central pore-forming subunit of HVA calcium channels.
Collapse
Affiliation(s)
- Alan Neely
- Centro Interdisciplinario de Neurociencia de Valparaíso and Facultad de Ciencias, Universidad de Valparaíso Valparaíso, Chile
| | - Patricia Hidalgo
- Forschungszentrum Jülich, Institute of Complex Systems 4, Zelluläre Biophysik Jülich, Germany
| |
Collapse
|
42
|
Hasreiter J, Goldnagl L, Böhm S, Kubista H. Cav1.2 and Cav1.3 L-type calcium channels operate in a similar voltage range but show different coupling to Ca(2+)-dependent conductances in hippocampal neurons. Am J Physiol Cell Physiol 2014; 306:C1200-13. [PMID: 24760982 DOI: 10.1152/ajpcell.00329.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the central nervous system, L-type voltage-gated calcium channels (LTCCs) come in two isoforms, namely Cav1.2 and Cav1.3 channels. It has been shown previously that these channels differ in biophysical properties, in subcellular localization, and in the coupling to the gene transcription machinery. In previous work on rat hippocampal neurons we have identified an excitatory cation conductance and an inhibitory potassium conductance as important LTCC coupling partners. Notably, a stimulus-dependent interplay of LTCC-mediated Ca(2+) influx and activation of these Ca(2+)-dependent conductances was found to give rise to characteristic voltage responses. However, the contribution of Cav1.2 and Cav1.3 to these voltage responses remained unknown. Hence, the relative contribution of the LTCC isoforms therein was the focus of the current study on hippocampal neurons derived from genetically modified mice, which either lack a LTCC isoform (Cav1.3 knockout mice) or express a dihydropyridine-insensitive LTCC isoform (Cav1.2DHP(-)-knockin mice). We identified common and alternate ion channel couplings of Cav1.2 and Cav1.3 channels. Whereas hyperpolarizing Ca(2+)-dependent conductances were coupled to both Cav1.2 and Cav1.3 channels, an afterdepolarizing potential was only induced by the activity of Cav1.2 channels. Unexpectedly, the activity of Cav1.2 channels was found at relatively hyperpolarized membrane voltages. Our data add important information about the differences between Cav1.2 and Cav1.3 channels that furthers our understanding of the physiological and pathophysiological neuronal roles of these calcium channels. Moreover, our findings suggest that Cav1.3 knockout mice together with Cav1.2DHP(-)-knockin mice provide valuable models for future investigation of hippocampal LTCC-dependent afterdepolarizations.
Collapse
Affiliation(s)
- Julia Hasreiter
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lena Goldnagl
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Böhm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
DuPont JJ, Hill MA, Bender SB, Jaisser F, Jaffe IZ. Aldosterone and vascular mineralocorticoid receptors: regulators of ion channels beyond the kidney. Hypertension 2014; 63:632-7. [PMID: 24379184 PMCID: PMC3954941 DOI: 10.1161/hypertensionaha.113.01273] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
44
|
Hofmann F, Flockerzi V, Kahl S, Wegener JW. L-type CaV1.2 calcium channels: from in vitro findings to in vivo function. Physiol Rev 2014; 94:303-26. [PMID: 24382889 DOI: 10.1152/physrev.00016.2013] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The L-type Cav1.2 calcium channel is present throughout the animal kingdom and is essential for some aspects of CNS function, cardiac and smooth muscle contractility, neuroendocrine regulation, and multiple other processes. The L-type CaV1.2 channel is built by up to four subunits; all subunits exist in various splice variants that potentially affect the biophysical and biological functions of the channel. Many of the CaV1.2 channel properties have been analyzed in heterologous expression systems including regulation of the L-type CaV1.2 channel by Ca(2+) itself and protein kinases. However, targeted mutations of the calcium channel genes confirmed only some of these in vitro findings. Substitution of the respective serines by alanine showed that β-adrenergic upregulation of the cardiac CaV1.2 channel did not depend on the phosphorylation of the in vitro specified amino acids. Moreover, well-established in vitro phosphorylation sites of the CaVβ2 subunit of the cardiac L-type CaV1.2 channel were found to be irrelevant for the in vivo regulation of the channel. However, the molecular basis of some kinetic properties, such as Ca(2+)-dependent inactivation and facilitation, has been approved by in vivo mutagenesis of the CaV1.2α1 gene. This article summarizes recent findings on the in vivo relevance of well-established in vitro results.
Collapse
|
45
|
Cardiac functions of voltage-gated Ca(2+) channels: role of the pharmacoresistant type (E-/R-Type) in cardiac modulation and putative implication in sudden unexpected death in epilepsy (SUDEP). Rev Physiol Biochem Pharmacol 2014; 167:115-39. [PMID: 25280639 DOI: 10.1007/112_2014_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Voltage-gated Ca(2+) channels (VGCCs) are ubiquitous in excitable cells. These channels play key roles in many physiological events like cardiac regulation/pacemaker activity due to intracellular Ca(2+) transients. In the myocardium, the Cav1 subfamily (L-type: Cav1.2 and Cav1.3) is the main contributor to excitation-contraction coupling and/or pacemaking, whereas the Cav3 subfamily (T-type: Cav3.1 and Cav3.2) is important in rhythmically firing of the cardiac nodal cells. No established cardiac function has been attributed to the Cav2 family (E-/R-type: Cav2.3) despite accumulating evidence of cardiac dysregulation observed upon deletion of the Cav2.3 gene, the only member of this family so far detected in cardiomyocytes. In this review, we summarize the pathophysiological changes observed after ablation of the E-/R-type VGCC and propose a cardiac mechanism of action for this channel. Also, considering the role played by this channel in epilepsy and its reported sensitivity to antiepileptic drugs, a putative involvement of this channel in the cardiac mechanism of sudden unexpected death in epilepsy is also discussed.
Collapse
|
46
|
HARTZELL HCRISS, DUCHATELLE-GOURDON ISABELLE. Structure and Neural Modulation of Cardiac Calcium Channels. J Cardiovasc Electrophysiol 2013. [DOI: 10.1111/j.1540-8167.1992.tb01937.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Felix R, Calderón-Rivera A, Andrade A. Regulation of high-voltage-activated Ca 2+ channel function, trafficking, and membrane stability by auxiliary subunits. ACTA ACUST UNITED AC 2013; 2:207-220. [PMID: 24949251 DOI: 10.1002/wmts.93] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Voltage-gated Ca2+ (CaV) channels mediate Ca2+ ions influx into cells in response to depolarization of the plasma membrane. They are responsible for initiation of excitation-contraction and excitation-secretion coupling, and the Ca2+ that enters cells through this pathway is also important in the regulation of protein phosphorylation, gene transcription, and many other intracellular events. Initial electrophysiological studies divided CaV channels into low-voltage-activated (LVA) and high-voltage-activated (HVA) channels. The HVA CaV channels were further subdivided into L, N, P/Q, and R-types which are oligomeric protein complexes composed of an ion-conducting CaVα1 subunit and auxiliary CaVα2δ, CaVβ, and CaVγ subunits. The functional consequences of the auxiliary subunits include altered functional and pharmacological properties of the channels as well as increased current densities. The latter observation suggests an important role of the auxiliary subunits in membrane trafficking of the CaVα1 subunit. This includes the mechanisms by which CaV channels are targeted to the plasma membrane and to appropriate regions within a given cell. Likewise, the auxiliary subunits seem to participate in the mechanisms that remove CaV channels from the plasma membrane for recycling and/or degradation. Diverse studies have provided important clues to the molecular mechanisms involved in the regulation of CaV channels by the auxiliary subunits, and the roles that these proteins could possibly play in channel targeting and membrane Stabilization.
Collapse
Affiliation(s)
- Ricardo Felix
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| | - Aida Calderón-Rivera
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| | - Arturo Andrade
- Department of Neuroscience, Brown University, Providence, RI, USA
| |
Collapse
|
48
|
Tyson JR, Snutch TP. Molecular nature of voltage‐gated calcium channels: structure and species comparison. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/wmts.91] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- John R. Tyson
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
| | - Terrance P. Snutch
- Michael Smith Laboratories University of British Columbia Vancouver BC Canada
| |
Collapse
|
49
|
The α2δ subunits of voltage-gated calcium channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013. [DOI: 10.1016/j.bbamem.2012.11.019] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Swarup V, Srivastava AK, Padma MV, Moganty RR. Quantitative Profiling and Identification of Plasma Proteins of Spinocerebellar Ataxia Type 2 Patients. NEURODEGENER DIS 2013; 12:199-206. [DOI: 10.1159/000346585] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 12/17/2012] [Indexed: 11/19/2022] Open
|