1
|
Miranda-Laferte E, Barkovits K, Rozanova S, Jordan N, Marcus K, Hidalgo P. The membrane-associated β2e-subunit of voltage-gated calcium channels translocates to the nucleus and regulates gene expression. Front Physiol 2025; 16:1555934. [PMID: 40297778 PMCID: PMC12034931 DOI: 10.3389/fphys.2025.1555934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
The β-subunit (Cavβ) is a central component of the voltage-gated calcium channel complex. It lacks transmembrane domains and exhibits both channel-related and non-related functions. Previous studies have shown that, in the absence of the Cavα1 pore-forming subunit, electrostatic interactions between the N-terminus of Cavβ2e and the plasma membrane mediate its anchoring to the cell surface. Here, we demonstrate that, upon phospholipase C activation, Cavβ2e dissociates from the plasma membrane and homogeneously distributes between the cytosol and the nucleus. Mutagenesis analysis identified critical residues in the N-terminus of the protein, including a stretch of positively charged amino acids and a dileucine motif, which serve as nuclear import and export signals, respectively. Fusion of the Cavβ2e N-terminus to a trimeric YFP chimeric construct shows that this segment suffices for nuclear shuttling. Thus, the N-terminus of Cavβ2e emerges as a regulatory hotspot region controlling the subcellular localization of the protein. Quantitative mass spectrometry analysis revealed that the heterologous expression of a nuclear-enriched Cavβ2e mutant regulates gene expression. Our findings demonstrate the presence of active nuclear localization signals in Cavβ2e that enables its nuclear targeting and regulation of protein expression. Furthermore, they establish the membrane-associated Cavβ2e as a novel signaling mediator within the phospholipase C cascade.
Collapse
Affiliation(s)
- Erick Miranda-Laferte
- Institute of Biological Information Processing (IBI-1)- Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
| | - Katalin Barkovits
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Svitlana Rozanova
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Nadine Jordan
- Institute of Biological Information Processing (IBI-1)- Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Patricia Hidalgo
- Institute of Biological Information Processing (IBI-1)- Molecular and Cellular Physiology, Forschungszentrum Jülich, Jülich, Germany
- Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
2
|
Oz S, Keren-Raifman T, Sharon T, Subramaniam S, Pallien T, Katz M, Tsemakhovich V, Sholokh A, Watad B, Tripathy DR, Sasson G, Chomsky-Hecht O, Vysochek L, Schulz-Christian M, Fecher-Trost C, Zühlke K, Bertinetti D, Herberg FW, Flockerzi V, Hirsch JA, Klussmann E, Weiss S, Dascal N. Tripartite interactions of PKA catalytic subunit and C-terminal domains of cardiac Ca 2+ channel may modulate its β-adrenergic regulation. BMC Biol 2024; 22:276. [PMID: 39609812 PMCID: PMC11603854 DOI: 10.1186/s12915-024-02076-9] [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: 11/01/2023] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The β-adrenergic augmentation of cardiac contraction, by increasing the conductivity of L-type voltage-gated CaV1.2 channels, is of great physiological and pathophysiological importance. Stimulation of β-adrenergic receptors (βAR) activates protein kinase A (PKA) through separation of regulatory (PKAR) from catalytic (PKAC) subunits. Free PKAC phosphorylates the inhibitory protein Rad, leading to increased Ca2+ influx. In cardiomyocytes, the core subunit of CaV1.2, CaV1.2α1, exists in two forms: full-length or truncated (lacking the distal C-terminus (dCT)). Signaling efficiency is believed to emanate from protein interactions within multimolecular complexes, such as anchoring PKA (via PKAR) to CaV1.2α1 by A-kinase anchoring proteins (AKAPs). However, AKAPs are inessential for βAR regulation of CaV1.2 in heterologous models, and their role in cardiomyocytes also remains unclear. RESULTS We show that PKAC interacts with CaV1.2α1 in heart and a heterologous model, independently of Rad, PKAR, or AKAPs. Studies with peptide array assays and purified recombinant proteins demonstrate direct binding of PKAC to two domains in CaV1.2α1-CT: the proximal and distal C-terminal regulatory domains (PCRD and DCRD), which also interact with each other. Data indicate both partial competition and possible simultaneous interaction of PCRD and DCRD with PKAC. The βAR regulation of CaV1.2α1 lacking dCT (which harbors DCRD) was preserved, but subtly altered, in a heterologous model, the Xenopus oocyte. CONCLUSIONS We discover direct interactions between PKAC and two domains in CaV1.2α1. We propose that these tripartite interactions, if present in vivo, may participate in organizing the multimolecular signaling complex and fine-tuning the βAR effect in cardiomyocytes.
Collapse
Affiliation(s)
- Shimrit Oz
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
- Department of Neuroscience, Faculty of Medicine, The Ruth and Bruce Rappaport, Haifa, 3109601, Israel
| | - Tal Keren-Raifman
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
| | - Tom Sharon
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
| | - Suraj Subramaniam
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997601, Israel
| | - Tamara Pallien
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Moshe Katz
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
| | - Vladimir Tsemakhovich
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
| | - Anastasiia Sholokh
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Baraa Watad
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
| | - Debi Ranjan Tripathy
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997601, Israel
- National Forensic Science University, Radhanagar, Agartala, Tripura, 799001, India
| | - Giorgia Sasson
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997601, Israel
| | - Orna Chomsky-Hecht
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997601, Israel
| | - Leonid Vysochek
- Heart Center, Sheba Medical Center, Ramat Gan, 5262000, Israel
| | - Maike Schulz-Christian
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Claudia Fecher-Trost
- Experimentelle Und Klinische Pharmakologie & Toxikologie, Universität Des Saarlandes, Homburg, 66421, Germany
| | - Kerstin Zühlke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniela Bertinetti
- Department of Biochemistry, University of Kassel, Heinrich-Plett-Str. 40, Kassel, 34132, Germany
| | - Friedrich W Herberg
- Department of Biochemistry, University of Kassel, Heinrich-Plett-Str. 40, Kassel, 34132, Germany
| | - Veit Flockerzi
- Experimentelle Und Klinische Pharmakologie & Toxikologie, Universität Des Saarlandes, Homburg, 66421, Germany
| | - Joel A Hirsch
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997601, Israel
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
| | - Sharon Weiss
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel.
| | - Nathan Dascal
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997601, Tel Aviv, Israel.
| |
Collapse
|
3
|
Zheng S, Ye L. Hemodynamic Melody of Postnatal Cardiac and Pulmonary Development in Children with Congenital Heart Diseases. BIOLOGY 2024; 13:234. [PMID: 38666846 PMCID: PMC11048247 DOI: 10.3390/biology13040234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Hemodynamics is the eternal theme of the circulatory system. Abnormal hemodynamics and cardiac and pulmonary development intertwine to form the most important features of children with congenital heart diseases (CHDs), thus determining these children's long-term quality of life. Here, we review the varieties of hemodynamic abnormalities that exist in children with CHDs, the recently developed neonatal rodent models of CHDs, and the inspirations these models have brought us in the areas of cardiomyocyte proliferation and maturation, as well as in alveolar development. Furthermore, current limitations, future directions, and clinical decision making based on these inspirations are highlighted. Understanding how CHD-associated hemodynamic scenarios shape postnatal heart and lung development may provide a novel path to improving the long-term quality of life of children with CHDs, transplantation of stem cell-derived cardiomyocytes, and cardiac regeneration.
Collapse
Affiliation(s)
- Sixie Zheng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| |
Collapse
|
4
|
Wardas B, Schneider JG, Klugbauer N, Flockerzi V, Beck A. Englerin A Inhibits T-Type Voltage-Gated Calcium Channels at Low Micromolar Concentrations. Mol Pharmacol 2023; 104:144-153. [PMID: 37399325 DOI: 10.1124/molpharm.122.000651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/05/2023] Open
Abstract
Englerin A (EA) is a potent agonist of tetrameric transient receptor potential canonical (TRPC) ion channels containing TRPC4 and TRPC5 subunits. TRPC proteins form cation channels that are activated by plasma membrane receptors. They convert extracellular signals such as angiotensin II into cellular responses, whereupon Na+ and Ca2+ influx and depolarization of the plasma membrane occur. Via depolarization, voltage-gated Ca2+ (CaV) channels can be activated, further increasing Ca2+ influx. We investigated the extent to which EA also affects the functions of CaV channels using the high-voltage-activated L-type Ca2+ channel CaV1.2 and the low-voltage-activated T-type Ca2+ channels CaV3.1, CaV3.2, and CaV3.3. After expression of cDNAs in human embryonic kidney (HEK293) cells, EA inhibited currents through all T-type channels at half-maximal inhibitory concentrations (IC50) of 7.5 to 10.3 μM. In zona glomerulosa cells of the adrenal gland, angiotensin II-induced elevation of cytoplasmic Ca2+ concentration leads to aldosterone release. We identified transcripts of low- and high-voltage-activated CaV channels and of TRPC1 and TRPC5 in the human adrenocortical (HAC15) zona glomerulosa cell line. Although no EA-induced TRPC activity was measurable, Ca2+ channel blockers distinguished T- and L-type Ca2+ currents. EA blocked 60% of the CaV current in HAC15 cells and T- and L-type channels analyzed at -30 mV and 10 mV were inhibited with IC50 values of 2.3 and 2.6 μM, respectively. Although the T-type blocker Z944 reduced basal and angiotensin II-induced 24-hour aldosterone release, EA was not effective. In summary, we show here that EA blocks CaV1.2 and T-type CaV channels at low-micromolar concentrations. SIGNIFICANCE STATEMENT: In this study we showed that englerin A (EA), a potent agonist of tetrameric transient receptor potential canonical (TRPC)4- or TRPC5-containing channels and currently under investigation to treat certain types of cancer, also inhibits the L-type voltage-gated Ca2+ (CaV) channel CaV1.2 and the T-type CaV channels CaV3.1, CaV3.2, and CaV3.3 channels at low micromolar concentrations.
Collapse
Affiliation(s)
- Barbara Wardas
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Jochen G Schneider
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Norbert Klugbauer
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie/PZMS, Universität des Saarlandes, Homburg, Germany (B.W., V.F., A.B.); Department of Internal Medicine II, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany (J.G.S.); Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg (J.G.S.); and Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany (N.K.)
| |
Collapse
|
5
|
Galow AM, Brenmoehl J, Hoeflich A. Synergistic effects of hormones on structural and functional maturation of cardiomyocytes and implications for heart regeneration. Cell Mol Life Sci 2023; 80:240. [PMID: 37541969 PMCID: PMC10403476 DOI: 10.1007/s00018-023-04894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
The limited endogenous regenerative capacity of the human heart renders cardiovascular diseases a major health threat, thus motivating intense research on in vitro heart cell generation and cell replacement therapies. However, so far, in vitro-generated cardiomyocytes share a rather fetal phenotype, limiting their utility for drug testing and cell-based heart repair. Various strategies to foster cellular maturation provide some success, but fully matured cardiomyocytes are still to be achieved. Today, several hormones are recognized for their effects on cardiomyocyte proliferation, differentiation, and function. Here, we will discuss how the endocrine system impacts cardiomyocyte maturation. After detailing which features characterize a mature phenotype, we will contemplate hormones most promising to induce such a phenotype, the routes of their action, and experimental evidence for their significance in this process. Due to their pleiotropic effects, hormones might be not only valuable to improve in vitro heart cell generation but also beneficial for in vivo heart regeneration. Accordingly, we will also contemplate how the presented hormones might be exploited for hormone-based regenerative therapies.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| |
Collapse
|
6
|
Gotthardt M, Badillo-Lisakowski V, Parikh VN, Ashley E, Furtado M, Carmo-Fonseca M, Schudy S, Meder B, Grosch M, Steinmetz L, Crocini C, Leinwand L. Cardiac splicing as a diagnostic and therapeutic target. Nat Rev Cardiol 2023; 20:517-530. [PMID: 36653465 DOI: 10.1038/s41569-022-00828-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/19/2023]
Abstract
Despite advances in therapeutics for heart failure and arrhythmias, a substantial proportion of patients with cardiomyopathy do not respond to interventions, indicating a need to identify novel modifiable myocardial pathobiology. Human genetic variation associated with severe forms of cardiomyopathy and arrhythmias has highlighted the crucial role of alternative splicing in myocardial health and disease, given that it determines which mature RNA transcripts drive the mechanical, structural, signalling and metabolic properties of the heart. In this Review, we discuss how the analysis of cardiac isoform expression has been facilitated by technical advances in multiomics and long-read and single-cell sequencing technologies. The resulting insights into the regulation of alternative splicing - including the identification of cardiac splice regulators as therapeutic targets and the development of a translational pipeline to evaluate splice modulators in human engineered heart tissue, animal models and clinical trials - provide a basis for improved diagnosis and therapy. Finally, we consider how the medical and scientific communities can benefit from facilitated acquisition and interpretation of splicing data towards improved clinical decision-making and patient care.
Collapse
Affiliation(s)
- Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research Partner Site Berlin), Berlin, Germany.
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Victor Badillo-Lisakowski
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research Partner Site Berlin), Berlin, Germany
| | - Victoria Nicole Parikh
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Euan Ashley
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Palo Alto, CA, USA
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Marta Furtado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sarah Schudy
- Institute for Cardiomyopathies, Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies, Department of Medicine III, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research Partner Site Heidelberg-Mannheim), Heidelberg, Germany
| | - Markus Grosch
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Lars Steinmetz
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Claudia Crocini
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Leslie Leinwand
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| |
Collapse
|
7
|
Seitter H, Obkircher J, Grabher P, Hartl J, Zanetti L, Lux UT, Fotakis G, Fernández-Quintero ML, Kaserer T, Koschak A. A novel calcium channel Cavβ 2 splice variant with unique properties predominates in the retina. J Biol Chem 2023; 299:102972. [PMID: 36738788 PMCID: PMC10074810 DOI: 10.1016/j.jbc.2023.102972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Cavβ subunits are essential for surface expression of voltage-gated calcium channel complexes and crucially modulate biophysical properties like voltage-dependent inactivation. Here, we describe the discovery and characterization of a novel Cavβ2 variant with distinct features that predominates in the retina. We determined spliced exons in retinal transcripts of the Cacnb2 gene, coding for Cavβ2, by RNA-Seq data analysis and quantitative PCR. We cloned a novel Cavβ2 splice variant from mouse retina, which we are calling β2i, and investigated biophysical properties of calcium currents with this variant in a heterologous expression system as well as its intrinsic membrane interaction when expressed alone. Our data showed that β2i predominated in the retina with expression in photoreceptors and bipolar cells. Furthermore, we observed that the β2i N-terminus exhibited an extraordinary concentration of hydrophobic residues, a distinct feature not seen in canonical variants. The biophysical properties resembled known membrane-associated variants, and β2i exhibited both a strong membrane association and a propensity for clustering, which depended on hydrophobic residues in its N-terminus. We considered available Cavβ structure data to elucidate potential mechanisms underlying the observed characteristics but resolved N-terminus structures were lacking and thus, precluded clear conclusions. With this description of a novel N-terminus variant of Cavβ2, we expand the scope of functional variation through N-terminal splicing with a distinct form of membrane attachment. Further investigation of the molecular mechanisms underlying the features of β2i could provide new angles on the way Cavβ subunits modulate Ca2+ channels at the plasma membrane.
Collapse
Affiliation(s)
- Hartwig Seitter
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria.
| | - Jana Obkircher
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Patricia Grabher
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Julia Hartl
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Lucia Zanetti
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Uwe Thorsten Lux
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georgios Fotakis
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Teresa Kaserer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
8
|
Siller A, Hofer NT, Tomagra G, Burkert N, Hess S, Benkert J, Gaifullina A, Spaich D, Duda J, Poetschke C, Vilusic K, Fritz EM, Schneider T, Kloppenburg P, Liss B, Carabelli V, Carbone E, Ortner NJ, Striessnig J. β2-subunit alternative splicing stabilizes Cav2.3 Ca 2+ channel activity during continuous midbrain dopamine neuron-like activity. eLife 2022; 11:e67464. [PMID: 35792082 PMCID: PMC9307272 DOI: 10.7554/elife.67464] [Citation(s) in RCA: 7] [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: 02/11/2021] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
In dopaminergic (DA) Substantia nigra (SN) neurons Cav2.3 R-type Ca2+-currents contribute to somatodendritic Ca2+-oscillations. This activity may contribute to the selective degeneration of these neurons in Parkinson's disease (PD) since Cav2.3-knockout is neuroprotective in a PD mouse model. Here, we show that in tsA-201-cells the membrane-anchored β2-splice variants β2a and β2e are required to stabilize Cav2.3 gating properties allowing sustained Cav2.3 availability during simulated pacemaking and enhanced Ca2+-currents during bursts. We confirmed the expression of β2a- and β2e-subunit transcripts in the mouse SN and in identified SN DA neurons. Patch-clamp recordings of mouse DA midbrain neurons in culture and SN DA neurons in brain slices revealed SNX-482-sensitive R-type Ca2+-currents with voltage-dependent gating properties that suggest modulation by β2a- and/or β2e-subunits. Thus, β-subunit alternative splicing may prevent a fraction of Cav2.3 channels from inactivation in continuously active, highly vulnerable SN DA neurons, thereby also supporting Ca2+ signals contributing to the (patho)physiological role of Cav2.3 channels in PD.
Collapse
Affiliation(s)
- Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Giulia Tomagra
- Department of Drug Science, NIS Centre, University of TorinoTorinoItaly
| | - Nicole Burkert
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Simon Hess
- Institute for Zoology, Biocenter, University of CologneCologneGermany
| | - Julia Benkert
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Aisylu Gaifullina
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Desiree Spaich
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | - Johanna Duda
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
| | | | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Toni Schneider
- Institute of Neurophysiology, University of CologneCologneGermany
| | - Peter Kloppenburg
- Institute for Zoology, Biocenter, University of CologneCologneGermany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, GermanyUlmGermany
- Linacre College & New College, University of OxfordOxfordUnited Kingdom
| | | | - Emilio Carbone
- Department of Drug Science, NIS Centre, University of TorinoTorinoItaly
| | - Nadine Jasmin Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of InnsbruckInnsbruckAustria
| |
Collapse
|
9
|
Metzner C, Mäki-Marttunen T, Karni G, McMahon-Cole H, Steuber V. The effect of alterations of schizophrenia-associated genes on gamma band oscillations. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:46. [PMID: 35854005 PMCID: PMC9261091 DOI: 10.1038/s41537-022-00255-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
Abnormalities in the synchronized oscillatory activity of neurons in general and, specifically in the gamma band, might play a crucial role in the pathophysiology of schizophrenia. While these changes in oscillatory activity have traditionally been linked to alterations at the synaptic level, we demonstrate here, using computational modeling, that common genetic variants of ion channels can contribute strongly to this effect. Our model of primary auditory cortex highlights multiple schizophrenia-associated genetic variants that reduce gamma power in an auditory steady-state response task. Furthermore, we show that combinations of several of these schizophrenia-associated variants can produce similar effects as the more traditionally considered synaptic changes. Overall, our study provides a mechanistic link between schizophrenia-associated common genetic variants, as identified by genome-wide association studies, and one of the most robust neurophysiological endophenotypes of schizophrenia.
Collapse
Affiliation(s)
- Christoph Metzner
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany.
- Biocomputation Research Group, School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom.
| | | | - Gili Karni
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany
- Minerva Schools at KGI, San Francisco, CA, USA
| | - Hana McMahon-Cole
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany
- Minerva Schools at KGI, San Francisco, CA, USA
| | - Volker Steuber
- Biocomputation Research Group, School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
10
|
Tani H, Tohyama S. Human Engineered Heart Tissue Models for Disease Modeling and Drug Discovery. Front Cell Dev Biol 2022; 10:855763. [PMID: 35433691 PMCID: PMC9008275 DOI: 10.3389/fcell.2022.855763] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/08/2022] [Indexed: 12/29/2022] Open
Abstract
The emergence of human induced pluripotent stem cells (hiPSCs) and efficient differentiation of hiPSC-derived cardiomyocytes (hiPSC-CMs) induced from diseased donors have the potential to recapitulate the molecular and functional features of the human heart. Although the immaturity of hiPSC-CMs, including the structure, gene expression, conduct, ion channel density, and Ca2+ kinetics, is a major challenge, various attempts to promote maturation have been effective. Three-dimensional cardiac models using hiPSC-CMs have achieved these functional and morphological maturations, and disease models using patient-specific hiPSC-CMs have furthered our understanding of the underlying mechanisms and effective therapies for diseases. Aside from the mechanisms of diseases and drug responses, hiPSC-CMs also have the potential to evaluate the safety and efficacy of drugs in a human context before a candidate drug enters the market and many phases of clinical trials. In fact, novel drug testing paradigms have suggested that these cells can be used to better predict the proarrhythmic risk of candidate drugs. In this review, we overview the current strategies of human engineered heart tissue models with a focus on major cardiac diseases and discuss perspectives and future directions for the real application of hiPSC-CMs and human engineered heart tissue for disease modeling, drug development, clinical trials, and cardiotoxicity tests.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
- *Correspondence: Shugo Tohyama,
| |
Collapse
|
11
|
IP3-dependent Ca2+ signals are tightly controlled by Cavβ3, but not by Cavβ1, 2 and 4. Cell Calcium 2022; 104:102573. [DOI: 10.1016/j.ceca.2022.102573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/17/2022] [Accepted: 03/15/2022] [Indexed: 11/18/2022]
|
12
|
Cruz-Garcia Y, Barkovits K, Kohlhaas M, Pickel S, Gulentz M, Heindl C, Pfeiffer K, Eder-Negrin P, Maack C, Marcus K, Kuhn M, Miranda-Laferte E. Nanoenviroments of the β-Subunit of L-Type Voltage-Gated Calcium Channels in Adult Cardiomyocytes. Front Cell Dev Biol 2022; 9:724778. [PMID: 35047492 PMCID: PMC8762238 DOI: 10.3389/fcell.2021.724778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
In cardiomyocytes, Ca2+ influx through L-type voltage-gated calcium channels (LTCCs) following membrane depolarization regulates crucial Ca2+-dependent processes including duration and amplitude of the action potentials and excitation-contraction coupling. LTCCs are heteromultimeric proteins composed of the Cavα1, Cavβ, Cavα2δ and Cavγ subunits. Here, using ascorbate peroxidase (APEX2)-mediated proximity labeling and quantitative proteomics, we identified 61 proteins in the nanoenvironments of Cavβ2 in cardiomyocytes. These proteins are involved in diverse cellular functions such as cellular trafficking, cardiac contraction, sarcomere organization and excitation-contraction coupling. Moreover, pull-down assays and co-immunoprecipitation analyses revealed that Cavβ2 interacts with the ryanodine receptor 2 (RyR2) in adult cardiomyocytes, probably coupling LTCCs and the RyR2 into a supramolecular complex at the dyads. This interaction is mediated by the Src-homology 3 domain of Cavβ2 and is necessary for an effective pacing frequency-dependent increase of the Ca2+-induced Ca2+ release mechanism in cardiomyocytes.
Collapse
Affiliation(s)
| | - Katalin Barkovits
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Michael Kohlhaas
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Simone Pickel
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Michelle Gulentz
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Cornelia Heindl
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Kathy Pfeiffer
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Petra Eder-Negrin
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany.,Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Erick Miranda-Laferte
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Institut für Biologische Informationsprozesse, Molekular- und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
13
|
Li J, Hua Y, Miyagawa S, Zhang J, Li L, Liu L, Sawa Y. hiPSC-Derived Cardiac Tissue for Disease Modeling and Drug Discovery. Int J Mol Sci 2020; 21:E8893. [PMID: 33255277 PMCID: PMC7727666 DOI: 10.3390/ijms21238893] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Relevant, predictive normal, or disease model systems are of vital importance for drug development. The difference between nonhuman models and humans could contribute to clinical trial failures despite ideal nonhuman results. As a potential substitute for animal models, human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) provide a powerful tool for drug toxicity screening, modeling cardiovascular diseases, and drug discovery. Here, we review recent hiPSC-CM disease models and discuss the features of hiPSC-CMs, including subtype and maturation and the tissue engineering technologies for drug assessment. Updates from the international multisite collaborators/administrations for development of novel drug discovery paradigms are also summarized.
Collapse
Affiliation(s)
- Junjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Cell Design for Tissue Construction, Faculty of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying Hua
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Jingbo Zhang
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Lingjun Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| | - Li Liu
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
- Department of Design for Tissue Regeneration, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (J.L.); (Y.H.); (S.M.); (J.Z.); (L.L.)
| |
Collapse
|
14
|
Regulation of cardiovascular calcium channel activity by post-translational modifications or interacting proteins. Pflugers Arch 2020; 472:653-667. [PMID: 32435990 DOI: 10.1007/s00424-020-02398-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
Abstract
Voltage-gated calcium channels are the major pathway for Ca2+ influx to initiate the contraction of smooth and cardiac muscles. Alterations of calcium channel function have been implicated in multiple cardiovascular diseases, such as hypertension, atrial fibrillation, and long QT syndrome. Post-translational modifications do expand cardiovascular calcium channel structure and function to affect processes such as channel trafficking or polyubiquitination by two E3 ubiquitin ligases, Ret finger protein 2 (Rfp2) or murine double minute 2 protein (Mdm2). Additionally, biophysical property such as Ca2+-dependent inactivation (CDI) could be altered through binding of calmodulin, or channel activity could be modulated via S-nitrosylation by nitric oxide and phosphorylation by protein kinases or by interacting protein partners, such as galectin-1 and Rem. Understanding how cardiovascular calcium channel function is post-translationally remodeled under distinctive disease conditions will provide better information about calcium channel-related disease mechanisms and improve the development of more selective therapeutic agents for cardiovascular diseases.
Collapse
|
15
|
Abstract
Maturation is the last phase of heart development that prepares the organ for strong, efficient, and persistent pumping throughout the mammal's lifespan. This process is characterized by structural, gene expression, metabolic, and functional specializations in cardiomyocytes as the heart transits from fetal to adult states. Cardiomyocyte maturation gained increased attention recently due to the maturation defects in pluripotent stem cell-derived cardiomyocyte, its antagonistic effect on myocardial regeneration, and its potential contribution to cardiac disease. Here, we review the major hallmarks of ventricular cardiomyocyte maturation and summarize key regulatory mechanisms that promote and coordinate these cellular events. With advances in the technical platforms used for cardiomyocyte maturation research, we expect significant progress in the future that will deepen our understanding of this process and lead to better maturation of pluripotent stem cell-derived cardiomyocyte and novel therapeutic strategies for heart disease.
Collapse
Affiliation(s)
- Yuxuan Guo
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - William Pu
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
16
|
Ortner NJ, Pinggera A, Hofer NT, Siller A, Brandt N, Raffeiner A, Vilusic K, Lang I, Blum K, Obermair GJ, Stefan E, Engel J, Striessnig J. RBP2 stabilizes slow Cav1.3 Ca 2+ channel inactivation properties of cochlear inner hair cells. Pflugers Arch 2019; 472:3-25. [PMID: 31848688 PMCID: PMC6960213 DOI: 10.1007/s00424-019-02338-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 12/04/2019] [Indexed: 01/31/2023]
Abstract
Cav1.3 L-type Ca2+ channels (LTCCs) in cochlear inner hair cells (IHCs) are essential for hearing as they convert sound-induced graded receptor potentials into tonic postsynaptic glutamate release. To enable fast and indefatigable presynaptic Ca2+ signaling, IHC Cav1.3 channels exhibit a negative activation voltage range and uniquely slow inactivation kinetics. Interaction with CaM-like Ca2+-binding proteins inhibits Ca2+-dependent inactivation, while the mechanisms underlying slow voltage-dependent inactivation (VDI) are not completely understood. Here we studied if the complex formation of Cav1.3 LTCCs with the presynaptic active zone proteins RIM2α and RIM-binding protein 2 (RBP2) can stabilize slow VDI. We detected both RIM2α and RBP isoforms in adult mouse IHCs, where they co-localized with Cav1.3 and synaptic ribbons. Using whole-cell patch-clamp recordings (tsA-201 cells), we assessed their effect on the VDI of the C-terminal full-length Cav1.3 (Cav1.3L) and a short splice variant (Cav1.342A) that lacks the C-terminal RBP2 interaction site. When co-expressed with the auxiliary β3 subunit, RIM2α alone (Cav1.342A) or RIM2α/RBP2 (Cav1.3L) reduced Cav1.3 VDI to a similar extent as observed in IHCs. Membrane-anchored β2 variants (β2a, β2e) that inhibit inactivation on their own allowed no further modulation of inactivation kinetics by RIM2α/RBP2. Moreover, association with RIM2α and/or RBP2 consolidated the negative Cav1.3 voltage operating range by shifting the channel's activation threshold toward more hyperpolarized potentials. Taken together, the association with "slow" β subunits (β2a, β2e) or presynaptic scaffolding proteins such as RIM2α and RBP2 stabilizes physiological gating properties of IHC Cav1.3 LTCCs in a splice variant-dependent manner ensuring proper IHC function.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Niels Brandt
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Andrea Raffeiner
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Isabelle Lang
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Gerald J Obermair
- Division of Physiology, Medical University Innsbruck, Innsbruck, Austria.,Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Eduard Stefan
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Jutta Engel
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
17
|
Mäki-Marttunen T, Devor A, Phillips WA, Dale AM, Andreassen OA, Einevoll GT. Computational Modeling of Genetic Contributions to Excitability and Neural Coding in Layer V Pyramidal Cells: Applications to Schizophrenia Pathology. Front Comput Neurosci 2019; 13:66. [PMID: 31616272 PMCID: PMC6775251 DOI: 10.3389/fncom.2019.00066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 09/09/2019] [Indexed: 11/13/2022] Open
Abstract
Pyramidal cells in layer V of the neocortex are one of the most widely studied neuron types in the mammalian brain. Due to their role as integrators of feedforward and cortical feedback inputs, they are well-positioned to contribute to the symptoms and pathology in mental disorders-such as schizophrenia-that are characterized by a mismatch between the internal perception and external inputs. In this modeling study, we analyze the input/output properties of layer V pyramidal cells and their sensitivity to modeled genetic variants in schizophrenia-associated genes. We show that the excitability of layer V pyramidal cells and the way they integrate inputs in space and time are altered by many types of variants in ion-channel and Ca2+ transporter-encoding genes that have been identified as risk genes by recent genome-wide association studies. We also show that the variability in the output patterns of spiking and Ca2+ transients in layer V pyramidal cells is altered by these model variants. Importantly, we show that many of the predicted effects are robust to noise and qualitatively similar across different computational models of layer V pyramidal cells. Our modeling framework reveals several aspects of single-neuron excitability that can be linked to known schizophrenia-related phenotypes and existing hypotheses on disease mechanisms. In particular, our models predict that single-cell steady-state firing rate is positively correlated with the coding capacity of the neuron and negatively correlated with the amplitude of a prepulse-mediated adaptation and sensitivity to coincidence of stimuli in the apical dendrite and the perisomatic region of a layer V pyramidal cell. These results help to uncover the voltage-gated ion-channel and Ca2+ transporter-associated genetic underpinnings of schizophrenia phenotypes and biomarkers.
Collapse
Affiliation(s)
| | - Anna Devor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.,Department of Radiology, University of California San Diego, La Jolla, CA, United States.,Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - William A Phillips
- Psychology, Faculty of Natural Sciences, University of Stirling, Stirling, United Kingdom
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.,Department of Radiology, University of California San Diego, La Jolla, CA, United States
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gaute T Einevoll
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
18
|
Mäki-Marttunen T, Krull F, Bettella F, Hagen E, Næss S, Ness TV, Moberget T, Elvsåshagen T, Metzner C, Devor A, Edwards AG, Fyhn M, Djurovic S, Dale AM, Andreassen OA, Einevoll GT. Alterations in Schizophrenia-Associated Genes Can Lead to Increased Power in Delta Oscillations. Cereb Cortex 2019; 29:875-891. [PMID: 30475994 PMCID: PMC6319172 DOI: 10.1093/cercor/bhy291] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/03/2018] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies have implicated many ion channels in schizophrenia pathophysiology. Although the functions of these channels are relatively well characterized by single-cell studies, the contributions of common variation in these channels to neurophysiological biomarkers and symptoms of schizophrenia remain elusive. Here, using computational modeling, we show that a common biomarker of schizophrenia, namely, an increase in delta-oscillation power, may be a direct consequence of altered expression or kinetics of voltage-gated ion channels or calcium transporters. Our model of a circuit of layer V pyramidal cells highlights multiple types of schizophrenia-related variants that contribute to altered dynamics in the delta-frequency band. Moreover, our model predicts that the same membrane mechanisms that increase the layer V pyramidal cell network gain and response to delta-frequency oscillations may also cause a deficit in a single-cell correlate of the prepulse inhibition, which is a behavioral biomarker highly associated with schizophrenia.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Simula Research Laboratory, Oslo, Norway
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Florian Krull
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Hagen
- Department of Physics, University of Oslo, Oslo, Norway
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Solveig Næss
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Torbjørn V Ness
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Torgeir Moberget
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Christoph Metzner
- Centre for Computer Science and Informatics Research, University of Hertfordshire, Hatfield, UK
| | - Anna Devor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Gaute T Einevoll
- Department of Physics, University of Oslo, Oslo, Norway
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
19
|
Conrad R, Stölting G, Hendriks J, Ruello G, Kortzak D, Jordan N, Gensch T, Hidalgo P. Rapid Turnover of the Cardiac L-Type Ca V1.2 Channel by Endocytic Recycling Regulates Its Cell Surface Availability. iScience 2018; 7:1-15. [PMID: 30267672 PMCID: PMC6135870 DOI: 10.1016/j.isci.2018.08.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/18/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Calcium entry through CaV1.2 L-type calcium channels regulates cardiac contractility. Here, we study the impact of exocytic and post-endocytic trafficking on cell surface channel abundance in cardiomyocytes. Single-molecule localization and confocal microscopy reveal an intracellular CaV1.2 pool tightly associated with microtubules from the perinuclear region to the cell periphery, and with actin filaments at the cell cortex. Channels newly inserted into the plasma membrane become internalized with an average time constant of 7.5 min and are sorted out to the Rab11a-recycling compartment. CaV1.2 recycling suffices for maintaining stable L-type current amplitudes over 20 hr independent of de novo channel transport along microtubules. Disruption of the actin cytoskeleton re-routes CaV1.2 from recycling toward lysosomal degradation. We identify endocytic recycling as essential for the homeostatic regulation of voltage-dependent calcium influx into cardiomyocytes. This mechanism provides the basis for a dynamic adjustment of the channel's surface availability and thus, of heart's contraction.
Collapse
Affiliation(s)
- Rachel Conrad
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Gabriel Stölting
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Johnny Hendriks
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Giovanna Ruello
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Daniel Kortzak
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Nadine Jordan
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Thomas Gensch
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Patricia Hidalgo
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany; Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
20
|
Sagawa H, Hoshino S, Yoshioka K, Ding WG, Omatsu-Kanbe M, Nakagawa M, Maruo Y, Matsuura H. Postnatal developmental changes in the sensitivity of L-type Ca 2+ channel to inhibition by verapamil in a mouse heart model. Pediatr Res 2018; 83:1207-1217. [PMID: 29554082 DOI: 10.1038/pr.2018.46] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/29/2018] [Indexed: 11/09/2022]
Abstract
BackgroundIn the clinical setting, verapamil is contraindicated in neonates and infants, because of the perceived risk of hypotension or bradyarrhythmia. However, it remains unclear whether there is an age-dependent difference in the sensitivity of cardiac L-type Ca2+ channel current (ICa,L) to inhibition by verapamil.MethodsVentricular myocytes were enzymatically dissociated from the hearts of six different age groups (0, 7, 14, 21, 28 days, and 10-15 weeks) of mice, using a similar Langendorff-perfusion method. Whole-cell patch-clamp technique was applied to examine the sensitivity of ICa,L to inhibition, by three classes of structurally different L-type Ca2+ channel antagonists.ResultsVerapamil, nifedipine, and diltiazem concentration-dependently blocked the ventricular ICa,L in all six age groups. However, although nifedipine and diltiazem blocked ventricular ICa,L with a similar potency in all age groups, verapamil more potently blocked ventricular ICa,L in day 0, day 7, day 14, and day 21 mice, than in day 28, and 10-15-week mice.ConclusionIn a mouse heart model, ventricular ICa,L before the weaning age (~21 days of age) exhibited a higher sensitivity to inhibition by verapamil than that after the weaning age, which may explain one possible mechanism associated with the development of verapamil-induced hypotension in human neonates and infants.
Collapse
Affiliation(s)
- Hironori Sagawa
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shinsuke Hoshino
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kengo Yoshioka
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Wei-Guang Ding
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Mariko Omatsu-Kanbe
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masao Nakagawa
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
21
|
Belkacemi A, Hui X, Wardas B, Laschke MW, Wissenbach U, Menger MD, Lipp P, Beck A, Flockerzi V. IP3 Receptor-Dependent Cytoplasmic Ca2+ Signals Are Tightly Controlled by Cavβ3. Cell Rep 2018; 22:1339-1349. [DOI: 10.1016/j.celrep.2018.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/10/2017] [Accepted: 01/03/2018] [Indexed: 12/11/2022] Open
|
22
|
Mäki-Marttunen T, Lines GT, Edwards AG, Tveito A, Dale AM, Einevoll GT, Andreassen OA. Pleiotropic effects of schizophrenia-associated genetic variants in neuron firing and cardiac pacemaking revealed by computational modeling. Transl Psychiatry 2017; 7:5. [PMID: 30446648 PMCID: PMC5802468 DOI: 10.1038/s41398-017-0007-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 01/01/2023] Open
Abstract
Schizophrenia patients have an increased risk of cardiac dysfunction. A possible factor underlying this comorbidity are the common variants in the large set of genes that have recently been discovered in genome-wide association studies (GWASs) as risk genes of schizophrenia. Many of these genes control the cell electrogenesis and calcium homeostasis. We applied biophysically detailed models of layer V pyramidal cells and sinoatrial node cells to study the contribution of schizophrenia-associated genes on cellular excitability. By including data from functional genomics literature to simulate the effects of common variants of these genes, we showed that variants of voltage-gated Na+ channel or hyperpolarization-activated cation channel-encoding genes cause qualitatively similar effects on layer V pyramidal cell and sinoatrial node cell excitability. By contrast, variants of Ca2+ channel or transporter-encoding genes mostly have opposite effects on cellular excitability in the two cell types. We also show that the variants may crucially affect the propagation of the cardiac action potential in the sinus node. These results may help explain some of the cardiac comorbidity in schizophrenia, and may facilitate generation of effective antipsychotic medications without cardiac side-effects such as arrhythmia.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway.
| | - Glenn T. Lines
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Aslak Tveito
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Anders M. Dale
- 0000 0001 2107 4242grid.266100.3Multimodal Imaging Laboratory, UC San Diego, La Jolla, CA USA ,0000 0001 2107 4242grid.266100.3Department of Neurosciences, University of California San Diego, La Jolla, CA USA ,0000 0001 2107 4242grid.266100.3Department of Radiology, University of California, San Diego, La Jolla, CA USA
| | - Gaute T. Einevoll
- 0000 0004 0607 975Xgrid.19477.3cDepartment of Mathematical Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway ,0000 0004 1936 8921grid.5510.1Department of Physics, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- 0000 0004 1936 8921grid.5510.1NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
23
|
Rosa N, Triffaux E, Robert V, Mars M, Klein M, Bouchaud G, Canivet A, Magnan A, Guéry JC, Pelletier L, Savignac M. The β and α2δ auxiliary subunits of voltage-gated calcium channel 1 (Ca v1) are required for T H2 lymphocyte function and acute allergic airway inflammation. J Allergy Clin Immunol 2017; 142:892-903.e8. [PMID: 29129580 DOI: 10.1016/j.jaci.2017.09.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/04/2017] [Accepted: 09/08/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND T lymphocytes express not only cell membrane ORAI calcium release-activated calcium modulator 1 but also voltage-gated calcium channel (Cav) 1 channels. In excitable cells these channels are composed of the ion-forming pore α1 and auxiliary subunits (β and α2δ) needed for proper trafficking and activation of the channel. Previously, we disclosed the role of Cav1.2 α1 in mouse and human TH2 but not TH1 cell functions and showed that knocking down Cav1 α1 prevents experimental asthma. OBJECTIVE We investigated the role of β and α2δ auxiliary subunits on Cav1 α1 function in TH2 lymphocytes and on the development of acute allergic airway inflammation. METHODS We used Cavβ antisense oligonucleotides to knock down Cavβ and gabapentin, a drug that binds to and inhibits α2δ1 and α2δ2, to test their effects on TH2 functions and their capacity to reduce allergic airway inflammation. RESULTS Mouse and human TH2 cells express mainly Cavβ1, β3, and α2δ2 subunits. Cavβ antisense reduces T-cell receptor-driven calcium responses and cytokine production by mouse and human TH2 cells with no effect on TH1 cells. Cavβ is mainly involved in restraining Cav1.2 α1 degradation through the proteasome because a proteasome inhibitor partially restores the α1 protein level. Gabapentin impairs the T-cell receptor-driven calcium response and cytokine production associated with the loss of α2δ2 protein in TH2 cells. CONCLUSIONS These results stress the role of Cavβ and α2δ2 auxiliary subunits in the stability and activation of Cav1.2 channels in TH2 lymphocytes both in vitro and in vivo, as demonstrated by the beneficial effect of Cavβ antisense and gabapentin in allergic airway inflammation.
Collapse
Affiliation(s)
- Nicolas Rosa
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France
| | - Emily Triffaux
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France
| | - Virginie Robert
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France
| | - Marion Mars
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France
| | - Martin Klein
- Institut du Thorax, INSERM CNRS, UNIV Nantes, France
| | | | - Astrid Canivet
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France
| | - Antoine Magnan
- Institut du Thorax, INSERM CNRS, UNIV Nantes, France; Centre Hospitalier Universitaire de Nantes, Service de Pneumologie, Nantes, France
| | - Jean-Charles Guéry
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France
| | - Lucette Pelletier
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France.
| | - Magali Savignac
- Center of Physiopathology Toulouse Purpan, University Paul Sabatier Toulouse III, INSERM U1043, CNRS UMR 5282, Toulouse, France.
| |
Collapse
|
24
|
Identification and prediction of alternative transcription start sites that generate rod photoreceptor-specific transcripts from ubiquitously expressed genes. PLoS One 2017. [PMID: 28640837 PMCID: PMC5480877 DOI: 10.1371/journal.pone.0179230] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Transcriptome complexity is substantially increased by the use of multiple transcription start sites for a given gene. By utilizing a rod photoreceptor-specific chromatin signature, and the RefSeq database of established transcription start sites, we have identified essentially all known rod photoreceptor genes as well as a group of novel genes that have a high probability of being expressed in rod photoreceptors. Approximately half of these novel rod genes are transcribed into multiple mRNA and/or protein isoforms through alternative transcriptional start sites (ATSS), only one of which has a rod-specific epigenetic signature and gives rise to a rod transcript. This suggests that, during retina development, some genes use ATSS to regulate cell type and temporal specificity, effectively generating a rod transcript from otherwise ubiquitously expressed genes. Biological confirmation of the relationship between epigenetic signatures and gene expression, as well as comparison of our genome-wide chromatin signature maps with available data sets for retina, namely a ChIP-on-Chip study of Polymerase-II (Pol-II) binding sites, ChIP-Seq studies for NRL- and CRX- binding sites and DHS (University of Washington data, available on UCSC mouse Genome Browser as a part of ENCODE project) fully support our hypothesis and together accurately identify and predict an array of new rod transcripts. The same approach was used to identify a number of TSS that are not currently in RefSeq. Biological conformation of the use of some of these TSS suggests that this method will be valuable for exploring the range of transcriptional complexity in many tissues. Comparison of mouse and human genome-wide data indicates that most of these alternate TSS appear to be present in both species, indicating that our approach can be useful for identification of regulatory regions that might play a role in human retinal disease.
Collapse
|
25
|
Oz S, Pankonien I, Belkacemi A, Flockerzi V, Klussmann E, Haase H, Dascal N. Protein kinase A regulates C-terminally truncated Ca V 1.2 in Xenopus oocytes: roles of N- and C-termini of the α 1C subunit. J Physiol 2017; 595:3181-3202. [PMID: 28194788 DOI: 10.1113/jp274015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/08/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS β-Adrenergic stimulation enhances Ca2+ entry via L-type CaV 1.2 channels, causing stronger contraction of cardiac muscle cells. The signalling pathway involves activation of protein kinase A (PKA), but the molecular details of PKA regulation of CaV 1.2 remain controversial despite extensive research. We show that PKA regulation of CaV 1.2 can be reconstituted in Xenopus oocytes when the distal C-terminus (dCT) of the main subunit, α1C , is truncated. The PKA upregulation of CaV 1.2 does not require key factors previously implicated in this mechanism: the clipped dCT, the A kinase-anchoring protein 15 (AKAP15), the phosphorylation sites S1700, T1704 and S1928, or the β subunit of CaV 1.2. The gating element within the initial segment of the N-terminus of the cardiac isoform of α1C is essential for the PKA effect. We propose that the regulation described here is one of two or several mechanisms that jointly mediate the PKA regulation of CaV 1.2 in the heart. ABSTRACT β-Adrenergic stimulation enhances Ca2+ currents via L-type, voltage-gated CaV 1.2 channels, strengthening cardiac contraction. The signalling via β-adrenergic receptors (β-ARs) involves elevation of cyclic AMP (cAMP) levels and activation of protein kinase A (PKA). However, how PKA affects the channel remains controversial. Recent studies in heterologous systems and genetically engineered mice stress the importance of the post-translational proteolytic truncation of the distal C-terminus (dCT) of the main (α1C ) subunit. Here, we successfully reconstituted the cAMP/PKA regulation of the dCT-truncated CaV 1.2 in Xenopus oocytes, which previously failed with the non-truncated α1C . cAMP and the purified catalytic subunit of PKA, PKA-CS, injected into intact oocytes, enhanced CaV 1.2 currents by ∼40% (rabbit α1C ) to ∼130% (mouse α1C ). PKA blockers were used to confirm specificity and the need for dissociation of the PKA holoenzyme. The regulation persisted in the absence of the clipped dCT (as a separate protein), the A kinase-anchoring protein AKAP15, and the phosphorylation sites S1700 and T1704, previously proposed as essential for the PKA effect. The CaV β2b subunit was not involved, as suggested by extensive mutagenesis. Using deletion/chimeric mutagenesis, we have identified the initial segment of the cardiac long-N-terminal isoform of α1C as a previously unrecognized essential element involved in PKA regulation. We propose that the observed regulation, that exclusively involves the α1C subunit, is one of several mechanisms underlying the overall PKA action on CaV 1.2 in the heart. We hypothesize that PKA is acting on CaV 1.2, in part, by affecting a structural 'scaffold' comprising the interacting cytosolic N- and C-termini of α1C .
Collapse
Affiliation(s)
- Shimrit Oz
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Ines Pankonien
- Max Delbrück Center for Molecular Medicine (MDC), D-13092, and the German Centre for Cardiovascular Research (DZHK) partner site, Berlin, Germany
| | - Anouar Belkacemi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421, Homburg, Germany
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421, Homburg, Germany
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine (MDC), D-13092, and the German Centre for Cardiovascular Research (DZHK) partner site, Berlin, Germany
| | - Hannelore Haase
- Max Delbrück Center for Molecular Medicine (MDC), D-13092, and the German Centre for Cardiovascular Research (DZHK) partner site, Berlin, Germany
| | - Nathan Dascal
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
26
|
Regulation of cardiac CACNB2 by microRNA-499: Potential role in atrial fibrillation. BBA CLINICAL 2017; 7:78-84. [PMID: 28239561 PMCID: PMC5318290 DOI: 10.1016/j.bbacli.2017.02.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/19/2022]
Abstract
The L-type calcium channel (LTCC) is one of the major ion channels that are known to be associated with the electrical remodeling of atrial fibrillation (AF). In AF, there is significant downregulation of the LTCC, but the underlying mechanism for such downregulation is not clear. We have previously reported that microRNA-499 (miR-499) is significantly upregulated in patients with permanent AF and that KCNN3, the gene that encodes the small-conductance calcium-activated potassium channel 3 (SK3), is a target of miR-499. We found that CACNB2, an important subunit of the LTCC, is also a target of miR-499. We hypothesize that miR-499 plays an important role in AF electrical remodeling by regulating the expression of CACNB2 and the LTCC. In atrial tissue from patients with permanent AF, CACNB2 was significantly downregulated by 67% (n = 4, p < 0.05) compared to those from patients with no history of AF. Transfection of miR-499 mimic into HL-1 cells, a mouse hyperplastic atrial cardiac myocyte cell-line, resulted in the downregulation of CACNB2 protein expression, while that of miR-499 inhibitor upregulated CACNB2 protein expression. Binding of miR-499 to the 3′ untranslated region of CACNB2 was confirmed by luciferase reporter assay and by the increased presence of CACNB2 mRNA in Argonaute pulled-down microRNA-induced silencing complexes after transfection with the miR-499 mimic. In addition, downregulation of CACNB2 resulted in the downregulation of protein levels of the pore-forming α-subunit (CACNA1C). In conclusion, upregulation of atrial miR-499 induces the downregulation of CACNB2 expression and may contribute to the electrical remodeling in AF. LTCC is downregulated with electrical remodeling of atrial fibrillation. MiR-499 is increased and CACNB2 is reduced in atria from patients with atrial fibrillation. MiR-499 binds to the 3′UTR of CACNB2 and inhibits its protein expression. Downregulation of CACNB2 results in the downregulation of LTCC pore-forming subunit. MiR-499 contributes to the electrical remodeling of AF through regulation of CACNB2.
Collapse
Key Words
- AF, atrial fibrillation
- Ago, Argonaute
- CACNA1C, voltage-dependent calcium channel α-1C subunit
- CACNB2, voltage-dependent calcium channel β-2 subunit
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- LTCC, L-type calcium channel
- SR, sinus rhythm
- UTR, untranslated region
- miR-499, microRNA-499
- miRISC, microRNA-inducing silencing complex
Collapse
|
27
|
Park CG, Park Y, Suh BC. The HOOK region of voltage-gated Ca2+ channel β subunits senses and transmits PIP2 signals to the gate. J Gen Physiol 2017; 149:261-276. [PMID: 28087621 PMCID: PMC5299622 DOI: 10.1085/jgp.201611677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/12/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022] Open
Abstract
The β subunit of voltage-gated Ca2+ (CaV) channels plays an important role in regulating gating of the α1 pore-forming subunit and its regulation by phosphatidylinositol 4,5-bisphosphate (PIP2). Subcellular localization of the CaV β subunit is critical for this effect; N-terminal-dependent membrane targeting of the β subunit slows inactivation and decreases PIP2 sensitivity. Here, we provide evidence that the HOOK region of the β subunit plays an important role in the regulation of CaV biophysics. Based on amino acid composition, we broadly divide the HOOK region into three domains: S (polyserine), A (polyacidic), and B (polybasic). We show that a β subunit containing only its A domain in the HOOK region increases inactivation kinetics and channel inhibition by PIP2 depletion, whereas a β subunit with only a B domain decreases these responses. When both the A and B domains are deleted, or when the entire HOOK region is deleted, the responses are elevated. Using a peptide-to-liposome binding assay and confocal microscopy, we find that the B domain of the HOOK region directly interacts with anionic phospholipids via polybasic and two hydrophobic Phe residues. The β2c-short subunit, which lacks an A domain and contains fewer basic amino acids and no Phe residues in the B domain, neither associates with phospholipids nor affects channel gating dynamically. Together, our data suggest that the flexible HOOK region of the β subunit acts as an important regulator of CaV channel gating via dynamic electrostatic and hydrophobic interaction with the plasma membrane.
Collapse
Affiliation(s)
- Cheon-Gyu Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
| | - Yongsoo Park
- Izmir International Biomedicine and Genome Institute (iBG-izmir), Dokuz Eylul University, 35340 Balcova, Izmir, Turkey
| | - Byung-Chang Suh
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
| |
Collapse
|
28
|
Tonegawa K, Otsuka W, Kumagai S, Matsunami S, Hayamizu N, Tanaka S, Moriwaki K, Obana M, Maeda M, Asahi M, Kiyonari H, Fujio Y, Nakayama H. Caveolae-specific activation loop between CaMKII and L-type Ca 2+ channel aggravates cardiac hypertrophy in α 1-adrenergic stimulation. Am J Physiol Heart Circ Physiol 2016; 312:H501-H514. [PMID: 28039202 DOI: 10.1152/ajpheart.00601.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 11/22/2022]
Abstract
Activation of CaMKII induces a myriad of biological processes and plays dominant roles in cardiac hypertrophy. Caveolar microdomain contains many calcium/calmodulin-dependent kinase II (CaMKII) targets, including L-type Ca2+ channel (LTCC) complex, and serves as a signaling platform. The location of CaMKII activation is thought to be critical; however, the roles of CaMKII in caveolae are still elusive due to lack of methodology for the assessment of caveolae-specific activation. Our aim was to develop a novel tool for the specific analysis of CaMKII activation in caveolae and to determine the functional role of caveolar CaMKII in cardiac hypertrophy. To assess the caveolae-specific activation of CaMKII, we generated a fusion protein composed of phospholamban and caveolin-3 (cPLN-Cav3) and GFP fusion protein with caveolin-binding domain fused to CaMKII inhibitory peptide (CBD-GFP-AIP), which inhibits CaMKII activation specifically in caveolae. Caveolae-specific activation of CaMKII was detected using phosphospecific antibody for PLN (Thr17). Furthermore, adenoviral overexpression of LTCC β2a-subunit (β2a) in NRCMs showed its constitutive phosphorylation by CaMKII, which induces hypertrophy, and that both phosphorylation and hypertrophy are abolished by CBD-GFP-AIP expression, indicating that β2a phosphorylation occurs specifically in caveolae. Finally, β2a phosphorylation was observed after phenylephrine stimulation in β2a-overexpressing mice, and attenuation of cardiac hypertrophy after chronic phenylephrine stimulation was observed in nonphosphorylated mutant of β2a-overexpressing mice. We developed novel tools for the evaluation and inhibition of caveolae-specific activation of CaMKII. We demonstrated that phosphorylated β2a dominantly localizes to caveolae and induces cardiac hypertrophy after α1-adrenergic stimulation in mice.NEW & NOTEWORTHY While signaling in caveolae is thought to be important in cardiac hypertrophy, direct evidence is missing due to lack of tools to assess caveolae-specific signaling. This is the first study to demonstrate caveolae-specific activation of CaMKII signaling in cardiac hypertrophy induced by α1-adrenergic stimulation using an originally developed tool.
Collapse
Affiliation(s)
- Kota Tonegawa
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Wataru Otsuka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Shohei Kumagai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Sachi Matsunami
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Nao Hayamizu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Kazumasa Moriwaki
- Faculty of Medicine, Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka, Japan; and
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Makiko Maeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Michio Asahi
- Faculty of Medicine, Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka, Japan; and
| | - Hiroshi Kiyonari
- Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan;
| |
Collapse
|
29
|
McKinnon D, Rosati B. Transmural gradients in ion channel and auxiliary subunit expression. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:165-186. [PMID: 27702655 DOI: 10.1016/j.pbiomolbio.2016.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
Evolution has acted to shape the action potential in different regions of the heart in order to produce a maximally stable and efficient pump. This has been achieved by creating regional differences in ion channel expression levels within the heart as well as differences between equivalent cardiac tissues in different species. These region- and species-dependent differences in channel expression are established by regulatory evolution, evolution of the regulatory mechanisms that control channel expression levels. Ion channel auxiliary subunits are obvious targets for regulatory evolution, in order to change channel expression levels and/or modify channel function. This review focuses on the transmural gradients of ion channel expression in the heart and the role that regulation of auxiliary subunit expression plays in generating and shaping these gradients.
Collapse
Affiliation(s)
- David McKinnon
- Department of Veterans Affairs Medical Center, Northport, NY, USA; Institute of Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Barbara Rosati
- Department of Veterans Affairs Medical Center, Northport, NY, USA; Institute of Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA; Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
30
|
Koch P, Herzig S, Matthes J. An expert protocol for immunofluorescent detection of calcium channels in tsA-201 cells. J Pharmacol Toxicol Methods 2016; 82:20-25. [PMID: 27421665 DOI: 10.1016/j.vascn.2016.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/08/2016] [Accepted: 07/11/2016] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Pore-forming subunits of voltage gated calcium channels (VGCC) are large membrane proteins (260kDa) containing 24 transmembrane domains. Despite transfection with viral promoter driven vectors, biochemical analysis of VGCC is often hampered by rather low expression levels in heterologous systems rendering VGCC challenging targets. Especially in immunofluorescent detection, calcium channels are demanding proteins. METHODS We provide an expert step-by-step protocol with adapted conditions for handling procedures (tsA-201 cell culture, transient transfection, incubation time and temperature at 28°C or 37°C and immunostaining) to address the L-type calcium-channel pore Cav1.2 in an immunofluorescent approach. RESULTS We performed immunocytochemical analysis of Cav1.2 expression at single-cell level in combination with detection of different markers for cellular organelles. We show confluency levels and shapes of tsA-201 cells at different time points during an experiment. Our experiments reveal sufficient levels of Cav1.2 protein and a correct Cav1.2 expression pattern in polygonal shaped cells already 12h after transfection. DISCUSSION A sequence of elaborated protocol modifications allows subcellular localization analysis of Cav1.2 in an immunocytochemical approach. We provide a protocol that may be used to achieve insights into physiological and pathophysiological processes involving voltage gated calcium channels. Our protocol may be used for expression analysis of other challenging proteins and efficient overexpression may be exploited in related biochemical techniques requiring immunolabels.
Collapse
Affiliation(s)
- Peter Koch
- Department of Pharmacology, University Clinic Cologne, Gleuelerstrasse 24, D-50931 Cologne, Germany.
| | - Stefan Herzig
- Department of Pharmacology, University Clinic Cologne, Gleuelerstrasse 24, D-50931 Cologne, Germany.
| | - Jan Matthes
- Department of Pharmacology, University Clinic Cologne, Gleuelerstrasse 24, D-50931 Cologne, Germany.
| |
Collapse
|
31
|
Liu J, Laksman Z, Backx PH. The electrophysiological development of cardiomyocytes. Adv Drug Deliv Rev 2016; 96:253-73. [PMID: 26788696 DOI: 10.1016/j.addr.2015.12.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/23/2015] [Accepted: 12/31/2015] [Indexed: 02/07/2023]
Abstract
The generation of human cardiomyocytes (CMs) from human pluripotent stem cells (hPSCs) has become an important resource for modeling human cardiac disease and for drug screening, and also holds significant potential for cardiac regeneration. Many challenges remain to be overcome however, before innovation in this field can translate into a change in the morbidity and mortality associated with heart disease. Of particular importance for the future application of this technology is an improved understanding of the electrophysiologic characteristics of CMs, so that better protocols can be developed and optimized for generating hPSC-CMs. Many different cell culture protocols are currently utilized to generate CMs from hPSCs and all appear to yield relatively “developmentally” immature CMs with highly heterogeneous electrical properties. These hPSC-CMs are characterized by spontaneous beating at highly variable rates with a broad range of depolarization-repolarization patterns, suggestive of mixed populations containing atrial, ventricular and nodal cells. Many recent studies have attempted to introduce approaches to promote maturation and to create cells with specific functional properties. In this review, we summarize the studies in which the electrical properties of CMs derived from stem cells have been examined. In order to place this information in a useful context, we also review the electrical properties of CMs as they transition from the developing embryo to the adult human heart. The signal pathways involved in the regulation of ion channel expression during development are also briefly considered.
Collapse
|
32
|
Mäki-Marttunen T, Halnes G, Devor A, Witoelar A, Bettella F, Djurovic S, Wang Y, Einevoll GT, Andreassen OA, Dale AM. Functional Effects of Schizophrenia-Linked Genetic Variants on Intrinsic Single-Neuron Excitability: A Modeling Study. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 1:49-59. [PMID: 26949748 DOI: 10.1016/j.bpsc.2015.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent genome-wide association studies have identified a large number of genetic risk factors for schizophrenia (SCZ) featuring ion channels and calcium transporters. For some of these risk factors, independent prior investigations have examined the effects of genetic alterations on the cellular electrical excitability and calcium homeostasis. In the present proof-of-concept study, we harnessed these experimental results for modeling of computational properties on layer V cortical pyramidal cells and identified possible common alterations in behavior across SCZ-related genes. METHODS We applied a biophysically detailed multicompartmental model to study the excitability of a layer V pyramidal cell. We reviewed the literature on functional genomics for variants of genes associated with SCZ and used changes in neuron model parameters to represent the effects of these variants. RESULTS We present and apply a framework for examining the effects of subtle single nucleotide polymorphisms in ion channel and calcium transporter-encoding genes on neuron excitability. Our analysis indicates that most of the considered SCZ-related genetic variants affect the spiking behavior and intracellular calcium dynamics resulting from summation of inputs across the dendritic tree. CONCLUSIONS Our results suggest that alteration in the ability of a single neuron to integrate the inputs and scale its excitability may constitute a fundamental mechanistic contributor to mental disease, alongside the previously proposed deficits in synaptic communication and network behavior.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Geir Halnes
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Anna Devor
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Aree Witoelar
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Francesco Bettella
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Yunpeng Wang
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Gaute T Einevoll
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| | - Anders M Dale
- Norwegian Centre for Mental Disorders Research and KG Jebsen Centre for Psychosis Research (TM-M, AW, FB, YW, OAA), Institute of Clinical Medicine, University of Oslo, Oslo; and Department of Mathematical Sciences and Technology (GH, GTE), Norwegian University of Life Sciences, Ås, Norway; Departments of Neurosciences (AD, YW, AMD) and Radiology (AD, AMD), University of California, San Diego, La Jolla, California; Martinos Center for Biomedical Imaging (AD), Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Division of Mental Health and Addiction (FB, YW, OAA) and Department of Medical Genetics (SD), Oslo University Hospital, Oslo; Norwegian Centre for Mental Disorders Research (SD), KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen; and Department of Physics (GTE), University of Oslo, Oslo, Norway
| |
Collapse
|
33
|
Moreno C, Hermosilla T, Morales D, Encina M, Torres-Díaz L, Díaz P, Sarmiento D, Simon F, Varela D. Cavβ2 transcription start site variants modulate calcium handling in newborn rat cardiomyocytes. Pflugers Arch 2015; 467:2473-84. [PMID: 26265381 DOI: 10.1007/s00424-015-1723-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/17/2015] [Indexed: 12/21/2022]
Abstract
In the heart, the main pathway for calcium influx is mediated by L-type calcium channels, a multi-subunit complex composed of the pore-forming subunit CaV1.2 and the auxiliary subunits CaVα2δ1 and CaVβ2. To date, five distinct CaVβ2 transcriptional start site (TSS) variants (CaVβ2a-e) varying only in the composition and length of the N-terminal domain have been described, each of them granting distinct biophysical properties to the L-type current. However, the physiological role of these variants in Ca(2+) handling in the native tissue has not been explored. Our results show that four of these variants are present in neonatal rat cardiomyocytes. The contribution of those CaVβ2 TSS variants on endogenous L-type current and Ca(2+) handling was explored by adenoviral-mediated overexpression of each CaVβ2 variant in cultured newborn rat cardiomyocytes. As expected, all CaVβ2 TSS variants increased L-type current density and produced distinctive changes on L-type calcium channel (LTCC) current activation and inactivation kinetics. The characteristics of the induced calcium transients were dependent on the TSS variant overexpressed. Moreover, the amplitude of the calcium transients varied depending on the subunit involved, being higher in cardiomyocytes transduced with CaVβ2a and smaller in CaVβ2d. Interestingly, the contribution of Ca(2+) influx and Ca(2+) release on total calcium transients, as well as the sarcoplasmic calcium content, was found to be TSS-variant-dependent. Remarkably, determination of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) messenger RNA (mRNA) abundance and cell size change indicates that CaVβ2 TSS variants modulate the cardiomyocyte hypertrophic state. In summary, we demonstrate that expression of individual CaVβ2 TSS variants regulates calcium handling in cardiomyocytes and, consequently, has significant repercussion in the development of hypertrophy.
Collapse
Affiliation(s)
- Cristian Moreno
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Tamara Hermosilla
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Danna Morales
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Matías Encina
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Leandro Torres-Díaz
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Pablo Díaz
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Daniela Sarmiento
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres Bello, Avenida Republica 239, Santiago, Chile
| | - Felipe Simon
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres Bello, Avenida Republica 239, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Varela
- Centro de Estudios Moleculares de la Célula (CEMC), Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
34
|
Kim DI, Park Y, Jang DJ, Suh BC. Dynamic phospholipid interaction of β2e subunit regulates the gating of voltage-gated Ca2+ channels. ACTA ACUST UNITED AC 2015; 145:529-41. [PMID: 25964431 PMCID: PMC4442786 DOI: 10.1085/jgp.201411349] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/10/2015] [Indexed: 12/18/2022]
Abstract
Membrane targeting of the β2e subunit is dynamically regulated by M1 muscarinic receptor signaling to promote fast inactivation of CaV2.2. High voltage-activated Ca2+ (CaV) channels are protein complexes containing pore-forming α1 and auxiliary β and α2δ subunits. The subcellular localization and membrane interactions of the β subunits play a crucial role in regulating CaV channel inactivation and its lipid sensitivity. Here, we investigated the effects of membrane phosphoinositide (PI) turnover on CaV2.2 channel function. The β2 isoform β2e associates with the membrane through electrostatic and hydrophobic interactions. Using chimeric β subunits and liposome-binding assays, we determined that interaction between the N-terminal 23 amino acids of β2e and anionic phospholipids was sufficient for β2e membrane targeting. Binding of the β2e subunit N terminus to liposomes was significantly increased by inclusion of 1% phosphatidylinositol 4,5-bisphosphate (PIP2) in the liposomes, suggesting that, in addition to phosphatidylserine, PIs are responsible for β2e targeting to the plasma membrane. Membrane binding of the β2e subunit slowed CaV2.2 current inactivation. When membrane phosphatidylinositol 4-phosphate and PIP2 were depleted by rapamycin-induced translocation of pseudojanin to the membrane, however, channel opening was decreased and fast inactivation of CaV2.2(β2e) currents was enhanced. Activation of the M1 muscarinic receptor elicited transient and reversible translocation of β2e subunits from membrane to cytosol, but not that of β2a or β3, resulting in fast inactivation of CaV2.2 channels with β2e. These results suggest that membrane targeting of the β2e subunit, which is mediated by nonspecific electrostatic insertion, is dynamically regulated by receptor stimulation, and that the reversible association of β2e with membrane PIs results in functional changes in CaV channel gating. The phospholipid–protein interaction observed here provides structural insight into mechanisms of membrane–protein association and the role of phospholipids in ion channel regulation.
Collapse
Affiliation(s)
- Dong-Il Kim
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Korea
| | - Yongsoo Park
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Deok-Jin Jang
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Kyungbuk 742-711, Korea
| | - Byung-Chang Suh
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Korea
| |
Collapse
|
35
|
Stölting G, de Oliveira RC, Guzman RE, Miranda-Laferte E, Conrad R, Jordan N, Schmidt S, Hendriks J, Gensch T, Hidalgo P. Direct interaction of CaVβ with actin up-regulates L-type calcium currents in HL-1 cardiomyocytes. J Biol Chem 2015; 290:4561-4572. [PMID: 25533460 PMCID: PMC4335199 DOI: 10.1074/jbc.m114.573956] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 12/05/2014] [Indexed: 12/14/2022] Open
Abstract
Expression of the β-subunit (CaVβ) is required for normal function of cardiac L-type calcium channels, and its up-regulation is associated with heart failure. CaVβ binds to the α1 pore-forming subunit of L-type channels and augments calcium current density by facilitating channel opening and increasing the number of channels in the plasma membrane, by a poorly understood mechanism. Actin, a key component of the intracellular trafficking machinery, interacts with Src homology 3 domains in different proteins. Although CaVβ encompasses a highly conserved Src homology 3 domain, association with actin has not yet been explored. Here, using co-sedimentation assays and FRET experiments, we uncover a direct interaction between CaVβ and actin filaments. Consistently, single-molecule localization analysis reveals streaklike structures composed by CaVβ2 that distribute over several micrometers along actin filaments in HL-1 cardiomyocytes. Overexpression of CaVβ2-N3 in HL-1 cells induces an increase in L-type current without altering voltage-dependent activation, thus reflecting an increased number of channels in the plasma membrane. CaVβ mediated L-type up-regulation, and CaVβ-actin association is prevented by disruption of the actin cytoskeleton with cytochalasin D. Our study reveals for the first time an interacting partner of CaVβ that is directly involved in vesicular trafficking. We propose a model in which CaVβ promotes anterograde trafficking of the L-type channels by anchoring them to actin filaments in their itinerary to the plasma membrane.
Collapse
Affiliation(s)
- Gabriel Stölting
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | | | - Raul E Guzman
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | - Erick Miranda-Laferte
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | - Rachel Conrad
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | - Nadine Jordan
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | - Silke Schmidt
- the Institut für Neurophysiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Johnny Hendriks
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | - Thomas Gensch
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and
| | - Patricia Hidalgo
- From the Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany and.
| |
Collapse
|
36
|
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
|
37
|
Wang DD, Bansal V, Fisher TE. The Ca2+ channel β2 subunit is selectively targeted to the axon terminals of supraoptic neurons. Channels (Austin) 2014; 8:216-21. [PMID: 24755552 DOI: 10.4161/chan.28863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The assembly of high voltage-activated Ca(2+) channels with different β subunits influences channel properties and possibly subcellular targeting. We studied β subunit expression in the somata and axon terminals of the magnocellular neurosecretory cells, which are located in the supraoptic nucleus (SON) and neurohypophysis, respectively. Antibodies directed against the 4 Ca(V)β subunits (Ca(V)β(1)-Ca(V)β(4)) were used for immunoblots and for immunostaining of slices of these two tissues. We found that all 4 β subunits are expressed in both locations, but that Ca(V)β(2) had the highest relative expression in the neurohypophysis. These data suggest that the Ca(V)β(2) subunit is selectively targeted to axon terminals and may play a role in targeting and/or regulating the properties of Ca(2+) channels.
Collapse
|
38
|
LUO HAIJIAN, SI JUNQIANG, ZHANG FENGJIE, YANG ZHENYU, WANG RUXING. Cardiac inotropic rebound effect after washout of acetylcholine is associated with electrophysiological heterogeneity in Langendorff-perfused rabbit heart. Exp Ther Med 2014; 7:755-757. [PMID: 24520282 PMCID: PMC3919906 DOI: 10.3892/etm.2014.1486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 01/09/2014] [Indexed: 11/15/2022] Open
Abstract
Cardiac electrophysiological heterogeneity related to the washout of acetylcholine (ACh) remains incompletely characterized. The aim of this study was to examine whether positive cardiac inotropic action is associated with electrophysiological heterogeneity between the atrium and the ventricle after ACh perfusion and washout. Epicardial monophasic action potentials (MAPs) from the right ventricle and right atrium, as well as cardiac contractility, were recorded from isolated Langendorff-perfused rabbit hearts using MAP electrodes and a force transducer. The results indicated that rebound positive inotropic actions were induced by ACh washout with adrenaline preconditioning. This effect was accompanied by an increase in MAP amplitude (MAPA) in the right ventricle but not the right atrium. These findings indicate that cholinergic muscarinic stimulation may lead to positive cardiac inotropic action followed by changes in regional electrophysiological heterogeneity between the atrial and ventricular myocardium. Therefore, we hypothesize that electrophysiological heterogeneity is an underlying cause of arrhythmogenesis as well as hemodynamic disturbance elicited by sudden termination of vagus stimulation.
Collapse
|
39
|
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
|
40
|
Miranda-Laferte E, Ewers D, Guzman RE, Jordan N, Schmidt S, Hidalgo P. The N-terminal domain tethers the voltage-gated calcium channel β2e-subunit to the plasma membrane via electrostatic and hydrophobic interactions. J Biol Chem 2014; 289:10387-10398. [PMID: 24519939 DOI: 10.1074/jbc.m113.507244] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The β-subunit associates with the α1 pore-forming subunit of high voltage-activated calcium channels and modulates several aspects of ion conduction. Four β-subunits are encoded by four different genes with multiple splice variants. Only two members of this family, β2a and β2e, associate with the plasma membrane in the absence of the α1-subunit. Palmitoylation on a di-cysteine motif located at the N terminus of β2a promotes membrane targeting and correlates with the unique ability of this protein to slow down inactivation. In contrast, the mechanism by which β2e anchors to the plasma membrane remains elusive. Here, we identified an N-terminal segment in β2e encompassing a cluster of positively charged residues, which is strictly required for membrane anchoring, and when transferred to the cytoplasmic β1b isoform it confers membrane localization to the latter. In the presence of negatively charged phospholipid vesicles, this segment binds to acidic liposomes dependently on the ionic strength, and the intrinsic fluorescence emission maxima of its single tryptophan blue shifts considerably. Simultaneous substitution of more than two basic residues impairs membrane targeting. Coexpression of the fast inactivating R-type calcium channels with wild-type β2e, but not with a β2e membrane association-deficient mutant, slows down inactivation. We propose that a predicted α-helix within this domain orienting parallel to the membrane tethers the β2e-subunit to the lipid bilayer via electrostatic interactions. Penetration of the tryptophan side chain into the lipidic core stabilizes the membrane-bound conformation. This constitutes a new mechanism for membrane anchoring among the β-subunit family that also sustains slowed inactivation.
Collapse
Affiliation(s)
- Erick Miranda-Laferte
- Institute of Complex Systems 4, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich
| | - David Ewers
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Raul E Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich
| | - Nadine Jordan
- Institute of Complex Systems 4, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich
| | - Silke Schmidt
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Patricia Hidalgo
- Institute of Complex Systems 4, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425 Jülich.
| |
Collapse
|
41
|
Striessnig J, Pinggera A, Kaur G, Bock G, Tuluc P. L-type Ca 2+ channels in heart and brain. ACTA ACUST UNITED AC 2014; 3:15-38. [PMID: 24683526 PMCID: PMC3968275 DOI: 10.1002/wmts.102] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
L-type calcium channels (Cav1) represent one of the three major classes (Cav1–3) of voltage-gated calcium channels. They were identified as the target of clinically used calcium channel blockers (CCBs; so-called calcium antagonists) and were the first class accessible to biochemical characterization. Four of the 10 known α1 subunits (Cav1.1–Cav1.4) form the pore of L-type calcium channels (LTCCs) and contain the high-affinity drug-binding sites for dihydropyridines and other chemical classes of organic CCBs. In essentially all electrically excitable cells one or more of these LTCC isoforms is expressed, and therefore it is not surprising that many body functions including muscle, brain, endocrine, and sensory function depend on proper LTCC activity. Gene knockouts and inherited human diseases have allowed detailed insight into the physiological and pathophysiological role of these channels. Genome-wide association studies and analysis of human genomes are currently providing even more hints that even small changes of channel expression or activity may be associated with disease, such as psychiatric disease or cardiac arrhythmias. Therefore, it is important to understand the structure–function relationship of LTCC isoforms, their differential contribution to physiological function, as well as their fine-tuning by modulatory cellular processes.
Collapse
Affiliation(s)
- Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Gurjot Kaur
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Gabriella Bock
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
42
|
Chernyavskaya Y, Ebert AM, Milligan E, Garrity DM. Voltage-gated calcium channel CACNB2 (β2.1) protein is required in the heart for control of cell proliferation and heart tube integrity. Dev Dyn 2012; 241:648-62. [DOI: 10.1002/dvdy.23746] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2012] [Indexed: 01/11/2023] Open
|
43
|
Gαi2- and Gαi3-specific regulation of voltage-dependent L-type calcium channels in cardiomyocytes. PLoS One 2011; 6:e24979. [PMID: 21966394 PMCID: PMC3180279 DOI: 10.1371/journal.pone.0024979] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 08/23/2011] [Indexed: 11/19/2022] Open
Abstract
Background Two pertussis toxin sensitive Gi proteins, Gi2 and Gi3, are expressed in cardiomyocytes and upregulated in heart failure. It has been proposed that the highly homologous Gi isoforms are functionally distinct. To test for isoform-specific functions of Gi proteins, we examined their role in the regulation of cardiac L-type voltage-dependent calcium channels (L-VDCC). Methods Ventricular tissues and isolated myocytes were obtained from mice with targeted deletion of either Gαi2 (Gαi2−/−) or Gαi3 (Gαi3−/−). mRNA levels of Gαi/o isoforms and L-VDCC subunits were quantified by real-time PCR. Gαi and Cavα1 protein levels as well as protein kinase B/Akt and extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation levels were assessed by immunoblot analysis. L-VDCC function was assessed by whole-cell and single-channel current recordings. Results In cardiac tissue from Gαi2−/− mice, Gαi3 mRNA and protein expression was upregulated to 187±21% and 567±59%, respectively. In Gαi3−/− mouse hearts, Gαi2 mRNA (127±5%) and protein (131±10%) levels were slightly enhanced. Interestingly, L-VDCC current density in cardiomyocytes from Gαi2−/− mice was lowered (−7.9±0.6 pA/pF, n = 11, p<0.05) compared to wild-type cells (−10.7±0.5 pA/pF, n = 22), whereas it was increased in myocytes from Gαi3−/− mice (−14.3±0.8 pA/pF, n = 14, p<0.05). Steady-state inactivation was shifted to negative potentials, and recovery kinetics slowed in the absence of Gαi2 (but not of Gαi3) and following treatment with pertussis toxin in Gαi3−/−. The pore forming Cavα1 protein level was unchanged in all mouse models analyzed, similar to mRNA levels of Cavα1 and Cavβ2 subunits. Interestingly, at the cellular signalling level, phosphorylation assays revealed abolished carbachol-triggered activation of ERK1/2 in mice lacking Gαi2. Conclusion Our data provide novel evidence for an isoform-specific modulation of L-VDCC by Gαi proteins. In particular, loss of Gαi2 is reflected by alterations in channel kinetics and likely involves an impairment of the ERK1/2 signalling pathway.
Collapse
|
44
|
Domes K, Ding J, Lemke T, Blaich A, Wegener JW, Brandmayr J, Moosmang S, Hofmann F. Truncation of murine CaV1.2 at Asp-1904 results in heart failure after birth. J Biol Chem 2011; 286:33863-71. [PMID: 21832054 DOI: 10.1074/jbc.m111.252312] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The carboxyl-terminal intracellular tail of the L-type Ca(2+) channel CaV1.2 modulates various aspects of channel activity.For example, deletion of the carboxyl-terminal sequence at Ser-1905 increased CaV1.2 currents in an expression model. To verify this finding in an animal model, we inserted three stop codons at the corresponding Asp-1904 in the murine CaV1.2 gene. Mice homozygous for the Stop mutation (Stop/Stop mice)were born at a Mendelian ratio but died after birth. Stop/Stop hearts showed reduced beating frequencies and contractions.Surprisingly, Stop/Stop cardiomyocytes displayed reduced IBa and a minor expression of the CaV1.2Stop protein. In contrast,expression of the CaV1.2Stop protein was normal in pooled smooth muscle samples from Stop/Stop embryos. As the CaV1.2 channel exists in a cardiac and smooth muscle splice variant, HK1 and LK1, respectively, we analyzed the consequences of the deletion of the carboxyl terminus in the respective splice variant using the rabbit CaV1.2 clone expressed in HEK293 cells.HEK293 cells transfected with the HK1Stop channel showed a reduced IBa and CaV1.2 expression. Treatment with proteasome inhibitors increased the expression of HK1Stop protein and IBa in HEK293 cells and in Stop/Stop cardiomyocytes indicating that truncation of CaV1.2 containing the cardiac exon 1a amino terminus results in proteasomal degradation of the translated protein. In contrast, HEK293 cells transfected with the LK1Stop channel had normal IBa and CaV1.2 expression. These findings indicate that absence of the carboxyl-terminal tail differentially determines the fate of the cardiac and smooth muscle splice variant of the CaV1.2 channel in the mouse.
Collapse
Affiliation(s)
- Katrin Domes
- From the Forschergruppe 923, Institut für Pharmakologie und Toxikologie, Technische Universität München, Biedersteiner Strasse 29, 80802 München, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Miranda-Laferte E, Gonzalez-Gutierrez G, Schmidt S, Zeug A, Ponimaskin EG, Neely A, Hidalgo P. Homodimerization of the Src homology 3 domain of the calcium channel β-subunit drives dynamin-dependent endocytosis. J Biol Chem 2011; 286:22203-10. [PMID: 21502319 DOI: 10.1074/jbc.m110.201871] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Voltage-dependent calcium channels constitute the main entry pathway for calcium into excitable cells. They are heteromultimers formed by an α(1) pore-forming subunit (Ca(V)α(1)) and accessory subunits. To achieve a precise coordination of calcium signals, the expression and activity of these channels is tightly controlled. The accessory β-subunit (Ca(V)β), a membrane associated guanylate kinase containing one guanylate kinase (β-GK) and one Src homology 3 (β-SH3) domain, has antagonistic effects on calcium currents by regulating different aspects of channel function. Although β-GK binds to a conserved site within the α(1)-pore-forming subunit and facilitates channel opening, β-SH3 binds to dynamin and promotes endocytosis. Here, we investigated the molecular switch underlying the functional duality of this modular protein. We show that β-SH3 homodimerizes through a single disulfide bond. Substitution of the only cysteine residue abolishes dimerization and impairs internalization of L-type Ca(V)1.2 channels expressed in Xenopus oocytes while preserving dynamin binding. Covalent linkage of the β-SH3 dimerization-deficient mutant yields a concatamer that binds to dynamin and restores endocytosis. Moreover, using FRET analysis, we show in living cells that Ca(V)β form oligomers and that this interaction is reduced by Ca(V)α(1). Association of Ca(V)β with a polypeptide encoding the binding motif in Ca(V)α(1) inhibited endocytosis. Together, these findings reveal that β-SH3 dimerization is crucial for endocytosis and suggest that channel activation and internalization are two mutually exclusive functions of Ca(V)β. We propose that a change in the oligomeric state of Ca(V)β is the functional switch between channel activator and channel internalizer.
Collapse
Affiliation(s)
- Erick Miranda-Laferte
- Institut für Neurophysiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Meissner M, Weissgerber P, Londoño JEC, Prenen J, Link S, Ruppenthal S, Molkentin JD, Lipp P, Nilius B, Freichel M, Flockerzi V. Moderate calcium channel dysfunction in adult mice with inducible cardiomyocyte-specific excision of the cacnb2 gene. J Biol Chem 2011; 286:15875-82. [PMID: 21357697 DOI: 10.1074/jbc.m111.227819] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The major L-type voltage-gated calcium channels in heart consist of an α1C (Ca(V)1.2) subunit usually associated with an auxiliary β subunit (Ca(V)β2). In embryonic cardiomyocytes, both the complete and the cardiac myocyte-specific null mutant of Ca(V)β2 resulted in reduction of L-type calcium currents by up to 75%, compromising heart function and causing defective remodeling of intra- and extra-embryonic blood vessels followed by embryonic death. Here we conditionally excised the Ca(V)β2 gene (cacnb2) specifically in cardiac myocytes of adult mice (KO). Upon gene deletion, Ca(V)β2 protein expression declined by >96% in isolated cardiac myocytes and by >74% in protein fractions from heart. These latter protein fractions include Ca(V)β2 proteins expressed in cardiac fibroblasts. Surprisingly, mice did not show any obvious impairment, although cacnb2 excision was not compensated by expression of other Ca(V)β proteins or changes of Ca(V)1.2 protein levels. Calcium currents were still dihydropyridine-sensitive, but current density at 0 mV was reduced by <29%. The voltage for half-maximal activation was slightly shifted to more depolarized potentials in KO cardiomyocytes when compared with control cells, but the difference was not significant. In summary, Ca(V)β2 appears to be a much stronger modulator of L-type calcium currents in embryonic than in adult cardiomyocytes. Although essential for embryonic survival, Ca(V)β2 down-regulation in cardiomyocytes is well tolerated by the adult mice.
Collapse
Affiliation(s)
- Marcel Meissner
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Facilitation of murine cardiac L-type Ca(v)1.2 channel is modulated by calmodulin kinase II-dependent phosphorylation of S1512 and S1570. Proc Natl Acad Sci U S A 2010; 107:10285-9. [PMID: 20479240 DOI: 10.1073/pnas.0914287107] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activity-dependent means of altering calcium (Ca(2)(+)) influx are assumed to be of great physiological consequence, although definitive tests of this assumption have only begun to emerge. Facilitation and inactivation offer two opposing, activity-dependent means of altering Ca(2+) influx via cardiac Ca(v)1.2 calcium channels. Voltage- and frequency-dependent facilitation of Ca(v)1.2 has been reported to depend on Calmodulin (CaM) and/or the activity of Calmodulin kinase II (CaMKII). Several sites within the cardiac L-type calcium channel complex have been proposed as the targets of CaMKII. Here, we generated mice with knockin mutations of alpha(1)1.2 S1512 and S1570 phosphorylation sites [sine facilitation (SF) mice]. Homocygote SF mice were viable and reproduced in a Mendelian ratio. Voltage-dependent facilitation in ventricular cardiomyocytes carrying the SF mutation was decreased from 1.58- to 1.18-fold. The CaMKII inhibitor KN-93 reduced facilitation to 1.28 in control cardiomyocytes. SF mutation negatively shifted the voltage-dependent inactivation and slowed recovery from inactivation, thereby making fewer channels available for activation. Telemetric ECG recordings at different heart rates showed that QT time decreased significantly more in SF than in control mice at higher rates. Our results strongly support the notion that CaMKII-dependent phosphorylation of Cav1.2 at S1512 and S1570 mediates Ca(2+) current facilitation in the murine heart.
Collapse
|