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
|
Bochereau P, Maman Haddad S, Le Bihan-Duval E, Berri C. RNA-Seq data provide new insights into the molecular regulation of breast muscle glycogen reserves, a key factor in muscle function and meat quality in chickens. Poult Sci 2025; 104:105136. [PMID: 40215882 PMCID: PMC12018111 DOI: 10.1016/j.psj.2025.105136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025] Open
Abstract
Research is needed to better understand the molecular mechanisms that influence muscle glycogen reserves in chickens due to their critical influence on muscle function and meat quality. In this study, breast muscle RNA sequencing data (RNA-Seq) were used to compare the transcriptomic profile of two original chicken lines divergently selected for breast muscle ultimate pH, which is a proxy for glycogen reserves. Weighted gene co-expression network analysis (WGCNA) of muscle and jejunum RNA-Seq data was also performed to highlight biological processes specifically involved in the gut-muscle dialogue that may contribute to the divergence in glycogen reserves between the two lines. Breast muscle RNA-Seq analysis of 4-week-old birds from the 15th generation of selection, in which glycogen reserves in the pHu- line were twice as high as that in the pHu+ line, revealed 2676 differentially expressed genes (Padj ≤ 0.05). Functional analysis of the genes overexpressed in the pHu- line highlighted enrichment in processes related to energy production from a wide range of substrates and pathways, as well as to processes involved in development of blood and lymph tissue. Diverse processes were enriched for genes overexpressed in the pHu+ line: muscle development and remodeling, lipid metabolism, immune response and inflammation, which suggested molecular changes much larger than those for carbohydrate metabolism. WGCNA revealed 64 modules of co-expressed genes. One, which contained 30 % genes expressed in the jejunum and 70 % genes expressed in the muscle, was correlated (P ≤ 0.05) with muscle glycogen reserves and several indicators of intestinal anatomy and health. Functional analysis of it showed an enrichment of processes related to transmission of nerve information and tissue oxygenation that seem to be involved in the gut-muscle dialogue that mediates establishment of breast muscle glycogen reserves. Finally, the study found that transcriptional regulations observed in muscle of the pHu+ line were similar to those in muscle afflicted with "wooden breast", which highlighted a dysfunction of mitochondrial metabolism and suggested several potential gene markers for both conditions.
Collapse
Affiliation(s)
| | - Sarah Maman Haddad
- SIGENAE, Université de Toulouse, GenPhyse, INRAE, ENVT, F-31326, Castanet Tolosan, France
| | | | - Cécile Berri
- INRAE, Université de Tours, BOA, 37380 Nouzilly, France.
| |
Collapse
|
3
|
Verbinnen I, Douzgou Houge S, Hsieh TC, Lesmann H, Kirchhoff A, Geneviève D, Brimble E, Lenaerts L, Haesen D, Levy RJ, Thevenon J, Faivre L, Marco E, Chong JX, Bamshad M, Patterson K, Mirzaa GM, Foss K, Dobyns W, White SM, Pais L, O'Heir E, Itzikowitz R, Donald KA, Van der Merwe C, Mussa A, Cervini R, Giorgio E, Roscioli T, Dias KR, Evans CA, Brown NJ, Ruiz A, Trujillo Quintero JP, Rabin R, Pappas J, Yuan H, Lachlan K, Thomas S, Devlin A, Wright M, Martin R, Karwowska J, Posmyk R, Chatron N, Stark Z, Heath O, Delatycki M, Buchert R, Korenke GC, Ramsey K, Narayanan V, Grange DK, Weisenberg JL, Haack TB, Karch S, Kipkemoi P, Mangi M, Bindels de Heus KGCB, de Wit MCY, Barakat TS, Lim D, Van Winckel G, Spillmann RC, Shashi V, Jacob M, Stehr AM, Krawitz P, Douzgos Houge G, Janssens V. Pathogenic de novo variants in PPP2R5C cause a neurodevelopmental disorder within the Houge-Janssens syndrome spectrum. Am J Hum Genet 2025; 112:554-571. [PMID: 39978342 PMCID: PMC11947181 DOI: 10.1016/j.ajhg.2025.01.021] [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: 08/27/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Pathogenic variants resulting in protein phosphatase 2A (PP2A) dysfunction result in mild to severe neurodevelopmental delay. PP2A is a trimer of a catalytic (C) subunit, scaffolding (A) subunit, and substrate binding/regulatory (B) subunit, encoded by 19 different genes. De novo missense variants in PPP2R5D (B56δ) or PPP2R1A (Aα) and de novo missense and loss-of-function variants in PPP2CA (Cα) lead to syndromes with overlapping phenotypic features, known as Houge-Janssens syndrome (HJS) types 1, 2, and 3, respectively. Here, we describe an additional condition in the HJS spectrum in 26 individuals with variants in PPP2R5C, encoding the regulatory B56γ subunit. Most changes were de novo and of the missense type. The clinical features were well within the HJS spectrum with strongest resemblance to HJS type 1, caused by B56δ variants. Common features were neurodevelopmental delay and hypotonia, with a high risk of epilepsy, behavioral problems, and mildly dysmorphic facial features. Head circumferences were above average or macrocephalic. The degree of intellectual disability was, on average, milder than in other HJS types. All variants affected either substrate binding (2/19), C-subunit binding (2/19), or both (15/19). Five variants were recurrent. Catalytic activity of the phosphatase was variably affected by the variants. Of note, PPP2R5C total loss-of-function variants could be inherited from a non-symptomatic parent. This implies that a dominant-negative mechanism on substrate dephosphorylation or general PP2A function is the most likely pathogenic mechanism.
Collapse
Affiliation(s)
- Iris Verbinnen
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium; KU Leuven Institute for Rare Diseases (Leuven.IRD), Leuven, Belgium
| | - Sofia Douzgou Houge
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tzung-Chien Hsieh
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Hellen Lesmann
- Institute of Human Genetics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Aron Kirchhoff
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - David Geneviève
- Montpellier University, INSERM U1183, Centre de Référence Anomalies du développement et syndromes malformatifs, ERN ITHACA, Génétique clinique, CHU Montpellier, Montpellier, France
| | | | - Lisa Lenaerts
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Dorien Haesen
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Rebecca J Levy
- Department of Neurology and Neurological Sciences, Stanford Medicine, Stanford, CA, USA
| | - Julien Thevenon
- CNRS UMR 5309, INSERM U1209, Institute of Advanced Biosciences, Université Grenoble-Alpes, Service Génomique et Procréation, Centre Hospitalo-Universitaire Grenoble Alpes, Cedex Grenoble, France
| | - Laurence Faivre
- Centre de génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'enfants, CHU Dijon Bourgogne, Dijon, France; UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France
| | | | - Jessica X Chong
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Mike Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Karynne Patterson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Ghayda M Mirzaa
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kimberly Foss
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William Dobyns
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Susan M White
- Victorian Clinical Genetics Services (VCGS), Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Lynn Pais
- Center for Mendelian Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emily O'Heir
- Center for Mendelian Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Raphaela Itzikowitz
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Celia Van der Merwe
- Stanley Center for Psychiatric Research, The Broad Institute, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Alessandro Mussa
- Department of Public Health and Pediatric Sciences, University of Torino, Regina Margherita Children's Hospital, Torino, Italy
| | - Raffaela Cervini
- Child Neuropsychiatry Department, Maria Vittoria Hospital, Torino, Italy
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS Mondino Foundation, Neurogenetics Research Centre, Pavia, Italy
| | - Tony Roscioli
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; Centre for Clinical Genetics, Sydney Children's Hospital, Sydney, NSW, Australia; New South Wales Health Pathology Randwick Genomics, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Kerith-Rae Dias
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2031, Australia
| | - Carey-Anne Evans
- Neuroscience Research Australia (NeuRA), Sydney, NSW, Australia; New South Wales Health Pathology Randwick Genomics, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| | - Natasha J Brown
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Anna Ruiz
- Genetics Laboratory, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, 08208 Sabadell, Spain
| | - Juan Pablo Trujillo Quintero
- Unitat de Genètica Clínica, Servei de Medicina Pediàtrica, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, 08208 Sabadell, Spain
| | - Rachel Rabin
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - John Pappas
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Hai Yuan
- Department of Pediatrics, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Katherine Lachlan
- Wessex Clinical Genetics Service, University Hospital Southampton, Princess Anne Hospital, Southampton SO16 5YA, UK
| | - Simon Thomas
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; Wessex Regional Genetics Laboratory, Salisbury NSF Foundation Trust, Salisbury District Hospital, Salisbury, UK
| | - Anita Devlin
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Richard Martin
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Institute of Genetic Medicine, Newcastle upon Tyne, UK
| | - Joanna Karwowska
- Department of Clinical Genetics, Medical University in Bialystok, Bialystok, Poland
| | - Renata Posmyk
- Department of Clinical Genetics, Medical University in Bialystok, Bialystok, Poland
| | - Nicolas Chatron
- Hospices Civils de Lyon, Groupe Hospitalier Est, Service de génétique, Bron, France; Université de Lyon, University Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, Lyon, France
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia; Australian Genomics Health Alliance, Melbourne, VIC, Australia; Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Oliver Heath
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia
| | - Martin Delatycki
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia; Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia; Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia
| | - Rebecca Buchert
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Georg-Christoph Korenke
- Klinik für Neuropädiatrie und angeborene Stoffwechselerkrankungen, Klinikum Oldenburg, Oldenburg, Germany
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Dorothy K Grange
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, One Children's Place, St. Louis, MO, USA
| | - Judith L Weisenberg
- Department of Pediatric Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Stephanie Karch
- Division of Pediatric Neurology and Metabolic Medicine, Department of Pediatrics I, Medical Faculty of Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Patricia Kipkemoi
- Neuroscience Unit, KEMRI-Wellcome Trust, Center for Geographic Medicine Research Coast, Kilifi, Kenya
| | - Moses Mangi
- Neuroscience Unit, KEMRI-Wellcome Trust, Center for Geographic Medicine Research Coast, Kilifi, Kenya
| | - Karen G C B Bindels de Heus
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, the Netherlands; ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Marie-Claire Y de Wit
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Neurology and Pediatric Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Tahsin Stefan Barakat
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Derek Lim
- Department of Clinical Genetics, Lavender House, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | | | - Rebecca C Spillmann
- Department of Pediatrics-Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Vandana Shashi
- Department of Pediatrics-Medical Genetics, Duke University School of Medicine, Durham, NC, USA
| | - Maureen Jacob
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Antonia M Stehr
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium; KU Leuven Institute for Rare Diseases (Leuven.IRD), Leuven, Belgium.
| |
Collapse
|
4
|
Carrillo ED, Alvarado JA, Hernández A, Lezama I, García MC, Sánchez JA. Thyroid Hormone Upregulates Cav1.2 Channels in Cardiac Cells via the Downregulation of the Channels' β4 Subunit. Int J Mol Sci 2024; 25:10798. [PMID: 39409130 PMCID: PMC11476369 DOI: 10.3390/ijms251910798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/27/2024] [Accepted: 10/06/2024] [Indexed: 10/20/2024] Open
Abstract
Thyroid hormone binds to specific nuclear receptors, regulating the expression of target genes, with major effects on cardiac function. Triiodothyronine (T3) increases the expression of key proteins related to calcium homeostasis, such as the sarcoplasmic reticulum calcium ATPase pump, but the detailed mechanism of gene regulation by T3 in cardiac voltage-gated calcium (Cav1.2) channels remains incompletely explored. Furthermore, the effects of T3 on Cav1.2 auxiliary subunits have not been investigated. We conducted quantitative reverse transcriptase polymerase chain reaction, Western blot, and immunofluorescence experiments in H9c2 cells derived from rat ventricular tissue, examining the effects of T3 on the expression of α1c, the principal subunit of Cav1.2 channels, and Cavβ4, an auxiliary Cav1.2 subunit that regulates gene expression. The translocation of phosphorylated cyclic adenosine monophosphate response element-binding protein (pCREB) by T3 was also examined. We found that T3 has opposite effects on these channel proteins, upregulating α1c and downregulating Cavβ4, and that it increases the nuclear translocation of pCREB while decreasing the translocation of Cavβ4. Finally, we found that overexpression of Cavβ4 represses the mRNA expression of α1c, suggesting that T3 upregulates the expression of the α1c subunit in response to a decrease in Cavβ4 subunit expression.
Collapse
Affiliation(s)
| | | | | | | | | | - Jorge A. Sánchez
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico; (E.D.C.); (J.A.A.); (A.H.); (I.L.); (M.C.G.)
| |
Collapse
|
5
|
Martus D, Williams SK, Pichi K, Mannebach-Götz S, Kaiser N, Wardas B, Fecher-Trost C, Meyer MR, Schmitz F, Beck A, Fairless R, Diem R, Flockerzi V, Belkacemi A. Cavβ3 Contributes to the Maintenance of the Blood-Brain Barrier and Alleviates Symptoms of Experimental Autoimmune Encephalomyelitis. Arterioscler Thromb Vasc Biol 2024; 44:1833-1851. [PMID: 38957986 DOI: 10.1161/atvbaha.124.321141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tight control of cytoplasmic Ca2+ concentration in endothelial cells is essential for the regulation of endothelial barrier function. Here, we investigated the role of Cavβ3, a subunit of voltage-gated Ca2+ (Cav) channels, in modulating Ca2+ signaling in brain microvascular endothelial cells (BMECs) and how this contributes to the integrity of the blood-brain barrier. METHODS We investigated the function of Cavβ3 in BMECs by Ca2+ imaging and Western blot, examined the endothelial barrier function in vitro and the integrity of the blood-brain barrier in vivo, and evaluated disease course after induction of experimental autoimmune encephalomyelitis in mice using Cavβ3-/- (Cavβ3-deficient) mice as controls. RESULTS We identified Cavβ3 protein in BMECs, but electrophysiological recordings did not reveal significant Cav channel activity. In vivo, blood-brain barrier integrity was reduced in the absence of Cavβ3. After induction of experimental autoimmune encephalomyelitis, Cavβ3-/- mice showed earlier disease onset with exacerbated clinical disability and increased T-cell infiltration. In vitro, the transendothelial resistance of Cavβ3-/- BMEC monolayers was lower than that of wild-type BMEC monolayers, and the organization of the junctional protein ZO-1 (zona occludens-1) was impaired. Thrombin stimulates inositol 1,4,5-trisphosphate-dependent Ca2+ release, which facilitates cell contraction and enhances endothelial barrier permeability via Ca2+-dependent phosphorylation of MLC (myosin light chain). These effects were more pronounced in Cavβ3-/- than in wild-type BMECs, whereas the differences were abolished in the presence of the MLCK (MLC kinase) inhibitor ML-7. Expression of Cacnb3 cDNA in Cavβ3-/- BMECs restored the wild-type phenotype. Coimmunoprecipitation and mass spectrometry demonstrated the association of Cavβ3 with inositol 1,4,5-trisphosphate receptor proteins. CONCLUSIONS Independent of its function as a subunit of Cav channels, Cavβ3 interacts with the inositol 1,4,5-trisphosphate receptor and is involved in the tight control of cytoplasmic Ca2+ concentration and Ca2+-dependent MLC phosphorylation in BMECs, and this role of Cavβ3 in BMECs contributes to blood-brain barrier integrity and attenuates the severity of experimental autoimmune encephalomyelitis disease.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Blood-Brain Barrier/metabolism
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium Channels/genetics
- Calcium Signaling
- Capillary Permeability
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Endothelial Cells/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Myosin Light Chains/metabolism
- Myosin-Light-Chain Kinase/metabolism
- Myosin-Light-Chain Kinase/genetics
- Phosphorylation
Collapse
Affiliation(s)
- Damian Martus
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Sarah K Williams
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Kira Pichi
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Stefanie Mannebach-Götz
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Nicolas Kaiser
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Barbara Wardas
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Claudia Fecher-Trost
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Markus R Meyer
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Frank Schmitz
- Institut für Anatomie und Zellbiologie (F.S.), Universität des Saarlandes, Homburg, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Richard Fairless
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (R.F., S.K.W.)
| | - Ricarda Diem
- Neurologische Klinik, Universitätsklinikum Heidelberg, Germany (S.K.W., K.P., R.F., R.D.)
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
| | - Anouar Belkacemi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Präklinisches Zentrum für Molekulare Signalverarbeitung, PharmaScienceHub (D.M., S.M.-G., N.K., B.W., C.F.-T., M.R.M., A. Beck, V.F., A. Belkacemi), Universität des Saarlandes, Homburg, Germany
- Now with Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Germany (A. Belkacemi)
| |
Collapse
|
6
|
Corzo-López A, Leyva-Leyva M, Castillo-Viveros V, Fernández-Gallardo M, Muñoz-Herrera D, Sandoval A, González-Ramírez R, Felix R. Molecular mechanisms of nuclear transport of the neuronal voltage-gated Ca 2+ channel β 3 auxiliary subunit. Neuroscience 2023:S0306-4522(23)00181-1. [PMID: 37169165 DOI: 10.1016/j.neuroscience.2023.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/01/2023] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
Previous studies have shown that in addition to its role within the voltage-gated calcium channel complex in the plasma membrane, the neuronal CaVβ subunit can translocate to the cell nucleus. However, little is known regarding the role this protein could play in the nucleus, nor the molecular mechanism used by CaVβ to enter this cell compartment. This report shows evidence that CaVβ3 has nuclear localization signals (NLS) that are not functional, suggesting that the protein does not use a classical nuclear import pathway. Instead, its entry into the nucleus could be associated with another protein that would function as a carrier, using a mechanism known as a piggyback. Mass spectrometry assays and bioinformatic analysis allowed the identification of proteins that could be participating in the entry of CaVβ3 into the nucleus. Likewise, through proximity ligation assays (PLA), it was found that members of the heterogeneous nuclear ribonucleoproteins (hnRNPs) and B56δ, a regulatory subunit of the protein phosphatase 2A (PP2A), could function as proteins that regulate this piggyback mechanism. On the other hand, bioinformatics and site-directed mutagenesis assays allowed the identification of a functional nuclear export signal (NES) that controls the exit of CaVβ3 from the nucleus, which would allow the completion of the nuclear transport cycle of the protein. These results reveal a novel mechanism for the nuclear transport cycle of the neuronal CaVβ3 subunit.
Collapse
Affiliation(s)
- Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Valeria Castillo-Viveros
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico; Department of Developmental Genetics and Molecular Physiology, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Mexico
| | | | - David Muñoz-Herrera
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| |
Collapse
|
7
|
Mayo S, Gómez-Manjón I, Marco-Hernández AV, Fernández-Martínez FJ, Camacho A, Martínez F. N-Type Ca Channel in Epileptic Syndromes and Epilepsy: A Systematic Review of Its Genetic Variants. Int J Mol Sci 2023; 24:6100. [PMID: 37047073 PMCID: PMC10094502 DOI: 10.3390/ijms24076100] [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: 02/20/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
N-type voltage-gated calcium channel controls the release of neurotransmitters from neurons. The association of other voltage-gated calcium channels with epilepsy is well-known. The association of N-type voltage-gated calcium channels and pain has also been established. However, the relationship between this type of calcium channel and epilepsy has not been specifically reviewed. Therefore, the present review systematically summarizes existing publications regarding the genetic associations between N-type voltage-dependent calcium channel and epilepsy.
Collapse
Affiliation(s)
- Sonia Mayo
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Irene Gómez-Manjón
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Ana Victoria Marco-Hernández
- Neuropediatric Unit, Hospital Universitario Doctor Peset, 46017 Valencia, Spain
- Translational Research in Genetics, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Francisco Javier Fernández-Martínez
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Ana Camacho
- Division of Pediatric Neurology, Department of Neurology, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Francisco Martínez
- Translational Research in Genetics, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Genomic Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politecnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
8
|
Oyama N, Vaneynde P, Reynhout S, Pao EM, Timms A, Fan X, Foss K, Derua R, Janssens V, Chung W, Mirzaa GM. Clinical, neuroimaging and molecular characteristics of PPP2R5D-related neurodevelopmental disorders: an expanded series with functional characterisation and genotype-phenotype analysis. J Med Genet 2022; 60:511-522. [PMID: 36216457 DOI: 10.1136/jmg-2022-108713] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/11/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Variants in PPP2R5D, affecting the regulatory B56δ subunit of protein phosphatase 2A (PP2A), have been identified in individuals with neurodevelopmental abnormalities. However, the molecular and clinical spectra remain incompletely understood. METHODS Individuals with PPP2R5D variants were enrolled through Simons Variation in Individuals Project/Simons Searchlight. Data were collected from medical history interviews, medical record review, online validated instruments and neuroimaging review. Genetic variants were biochemically characterised. RESULTS We studied 76 individuals with PPP2R5D variants, including 68 with pathogenic de novo variants, four with a variant of uncertain significance (VUS) and four siblings with a novel dominantly inherited pathogenic variant. Among 13 pathogenic variants, eight were novel and two (p.Glu198Lys and p.Glu200Lys) were highly recurrent. Functional analysis revealed impaired PP2A A/C-subunit binding, decreased short linear interaction motif-dependent substrate binding or both-with the most severe phenotypes associated with variants that completely retained one of these binding characteristics and lost the other-further supporting a dominant-negative disease mechanism. p.Glu198Lys showed the highest C-binding defect and a more severe clinical phenotype. The inherited p.Glu197Gly variant had a mild substrate binding defect, and three of four VUS had no biochemical impact. Common clinical phenotypes were language, intellectual or learning disabilities (80.6%), hypotonia (75.0%), macrocephaly (66.7%), seizures (45.8%) and autism spectrum disorder (26.4%). The mean composite Vineland score was 59.8, and most participants were in the 'moderate to low' and 'low' adaptive levels in all domains. CONCLUSION Our study delineates the most common features of PPP2R5D-related neurodevelopmental disorders, expands the clinical and molecular spectrum and identifies genotype-phenotype correlations.
Collapse
Affiliation(s)
- Nora Oyama
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Pieter Vaneynde
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Sara Reynhout
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Emily M Pao
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Andrew Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Xiao Fan
- Department of Pediatrics, Columbia University, New York City, New York, USA
| | - Kimberly Foss
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,SyBioMa, University of Leuven (KU Leuven), Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium.,KU Leuven Brain Institute (LBI), Leuven, Belgium
| | - Wendy Chung
- Department of Pediatrics, Columbia University, New York City, New York, USA.,Department of Medicine, Columbia University, New York City, New York, USA
| | - Ghayda M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA .,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
9
|
Hodges SL, Bouza AA, Isom LL. Therapeutic Potential of Targeting Regulated Intramembrane Proteolysis Mechanisms of Voltage-Gated Ion Channel Subunits and Cell Adhesion Molecules. Pharmacol Rev 2022; 74:1028-1048. [PMID: 36113879 PMCID: PMC9553118 DOI: 10.1124/pharmrev.121.000340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 05/13/2022] [Indexed: 10/03/2023] Open
Abstract
Several integral membrane proteins undergo regulated intramembrane proteolysis (RIP), a tightly controlled process through which cells transmit information across and between intracellular compartments. RIP generates biologically active peptides by a series of proteolytic cleavage events carried out by two primary groups of enzymes: sheddases and intramembrane-cleaving proteases (iCLiPs). Following RIP, fragments of both pore-forming and non-pore-forming ion channel subunits, as well as immunoglobulin super family (IgSF) members, have been shown to translocate to the nucleus to function in transcriptional regulation. As an example, the voltage-gated sodium channel β1 subunit, which is also an IgSF-cell adhesion molecule (CAM), is a substrate for RIP. β1 RIP results in generation of a soluble intracellular domain, which can regulate gene expression in the nucleus. In this review, we discuss the proposed RIP mechanisms of voltage-gated sodium, potassium, and calcium channel subunits as well as the roles of their generated proteolytic products in the nucleus. We also discuss other RIP substrates that are cleaved by similar sheddases and iCLiPs, such as IgSF macromolecules, including CAMs, whose proteolytically generated fragments function in the nucleus. Importantly, dysfunctional RIP mechanisms are linked to human disease. Thus, we will also review how understanding RIP events and subsequent signaling processes involving ion channel subunits and IgSF proteins may lead to the discovery of novel therapeutic targets. SIGNIFICANCE STATEMENT: Several ion channel subunits and immunoglobulin superfamily molecules have been identified as substrates of regulated intramembrane proteolysis (RIP). This signal transduction mechanism, which generates polypeptide fragments that translocate to the nucleus, is an important regulator of gene transcription. RIP may impact diseases of excitability, including epilepsy, cardiac arrhythmia, and sudden death syndromes. A thorough understanding of the role of RIP in gene regulation is critical as it may reveal novel therapeutic strategies for the treatment of previously intractable diseases.
Collapse
Affiliation(s)
- Samantha L Hodges
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Alexandra A Bouza
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Lori L Isom
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
10
|
Abstract
Voltage-gated Ca2+ (Cav) channels play pivotal roles in regulating gene transcription, neuronal excitability, and neurotransmitter release. To meet the spatial and temporal demands of visual signaling, Cav channels exhibit unusual properties in the retina compared to their counterparts in other areas of the nervous system. In this article, we review current concepts regarding the specific subtypes of Cav channels expressed in the retina, their intrinsic properties and forms of modulation, and how their dysregulation could lead to retinal disease.
Collapse
Affiliation(s)
- Brittany Williams
- Department of Cell Biology & Physiology, Carolina Institute for Developmental Disabilities, and Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - J Wesley Maddox
- Department of Neuroscience, University of Texas, Austin, Texas, USA;
| | - Amy Lee
- Department of Neuroscience, University of Texas, Austin, Texas, USA;
| |
Collapse
|
11
|
Vergnol A, Traoré M, Pietri-Rouxel F, Falcone S. New Insights in CaVβ Subunits: Role in the Regulation of Gene Expression and Cellular Homeostasis. Front Cell Dev Biol 2022; 10:880441. [PMID: 35465309 PMCID: PMC9019481 DOI: 10.3389/fcell.2022.880441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
The voltage-gated calcium channels (CaVs or VGCCs) are fundamental regulators of intracellular calcium homeostasis. When electrical activity induces their activation, the influx of calcium that they mediate or their interaction with intracellular players leads to changes in intracellular Ca2+ levels which regulate many processes such as contraction, secretion and gene expression, depending on the cell type. The essential component of the pore channel is the CaVα1 subunit. However, the fine-tuning of Ca2+-dependent signals is guaranteed by the modulatory role of the auxiliary subunits β, α2δ, and γ of the CaVs. In particular, four different CaVβ proteins (CaVβ1, CaVβ2, CaVβ3, and CaVβ4) are encoded by four different genes in mammalians, each of them displaying several splice variants. Some of these isoforms have been described in regulating CaVα1 docking and stability at the membrane and controlling the channel complex’s conformational changes. In addition, emerging evidences have highlighted other properties of the CaVβ subunits, independently of α1 and non-correlated to its channel or voltage sensing functions. This review summarizes the recent findings reporting novel roles of the auxiliary CaVβ subunits and in particular their direct or indirect implication in regulating gene expression in different cellular contexts.
Collapse
|
12
|
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]
|
13
|
Madaan P, Kaur A, Saini L, Paria P, Vyas S, Sharma AR, Sahu JK. PPP2R5D-Related Neurodevelopmental Disorder or Developmental and Epileptic Encephalopathy?: A Novel Phenotypic Description and Review of Published Cases. Neuropediatrics 2022; 53:20-25. [PMID: 34448180 DOI: 10.1055/s-0041-1733984] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Protein phosphatase 2 regulatory subunit B' delta (PPP2R5D)-related neurodevelopmental disorder is caused by pathogenic variations in the PPP2R5D gene, product of which is involved in dephosphorylation. This is a rare disorder with description limited to case reports. Its phenotypic spectrum has expanded over the last decade. METHODS We report a child with a developmental and epileptic encephalopathy phenotype with a pathogenic PPP2R5D variant. This phenotype has not been previously reported. We also reviewed the previously published reports of patients with this disorder. RESULTS Including the index child, 28 cases (15 girls) were identified from nine relevant research items for analysis. All patients had developmental delay. History of seizures was observed in seven patients while macrocephaly was seen in nearly 80% of patients. Nonneurological manifestations were observed in 13 patients with the most common one being ophthalmological manifestations. The most common genetic variation was c.G592A (p.E198K). The common phenotypic associations of this variation were developmental delay, macrocephaly (11/15), and epilepsy (6/15). CONCLUSION PPP2R5D gene variations should be suspected in children with developmental delay, autistic features, macrocephaly with or without epilepsy in the absence of any clear etiology. Dysmorphic features might provide a diagnostic clue. DEE phenotype may also be the presenting feature and might be an underreported entity.
Collapse
Affiliation(s)
- Priyanka Madaan
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amrit Kaur
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Lokesh Saini
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.,Department of Pediatrics, All India Institute of Medical Sciences, Jodhpur, India
| | - Pradip Paria
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sameer Vyas
- Department of Radiodiagnosis and Imaging (Section of Neuroimaging and Interventional Radiology), Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amit R Sharma
- Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jitendra K Sahu
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
14
|
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
|
15
|
Heck J, Palmeira Do Amaral AC, Weißbach S, El Khallouqi A, Bikbaev A, Heine M. More than a pore: How voltage-gated calcium channels act on different levels of neuronal communication regulation. Channels (Austin) 2021; 15:322-338. [PMID: 34107849 PMCID: PMC8205089 DOI: 10.1080/19336950.2021.1900024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) represent key regulators of the calcium influx through the plasma membrane of excitable cells, like neurons. Activated by the depolarization of the membrane, the opening of VGCCs induces very transient and local changes in the intracellular calcium concentration, known as calcium nanodomains, that in turn trigger calcium-dependent signaling cascades and the release of chemical neurotransmitters. Based on their central importance as concierges of excitation-secretion coupling and therefore neuronal communication, VGCCs have been studied in multiple aspects of neuronal function and malfunction. However, studies on molecular interaction partners and recent progress in omics technologies have extended the actual concept of these molecules. With this review, we want to illustrate some new perspectives of VGCCs reaching beyond their function as calcium-permeable pores in the plasma membrane. Therefore, we will discuss the relevance of VGCCs as voltage sensors in functional complexes with ryanodine receptors, channel-independent actions of auxiliary VGCC subunits, and provide an insight into how VGCCs even directly participate in gene regulation. Furthermore, we will illustrate how structural changes in the intracellular C-terminus of VGCCs generated by alternative splicing events might not only affect the biophysical channel characteristics but rather determine their molecular environment and downstream signaling pathways.
Collapse
Affiliation(s)
- Jennifer Heck
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Ana Carolina Palmeira Do Amaral
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Stephan Weißbach
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
- Computational Genomics and Bioinformatics, Johannes Gutenberg-University Mainz, University Medical Center Mainz, Institute for Human Genetics, Mainz, Germany
| | - Abderazzaq El Khallouqi
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Arthur Bikbaev
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Martin Heine
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| |
Collapse
|
16
|
Jia Y, Wang X, Chen Y, Qiu W, Ge W, Ma C. Proteomic and Transcriptomic Analyses Reveal Pathological Changes in the Entorhinal Cortex Region that Correlate Well with Dysregulation of Ion Transport in Patients with Alzheimer's Disease. Mol Neurobiol 2021; 58:4007-4027. [PMID: 33904022 DOI: 10.1007/s12035-021-02356-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/10/2021] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. The earliest neuropathology of AD appears in entorhinal cortex (EC) regions. Therapeutic strategies and preventive measures to protect against entorhinal degeneration would be of substantial value in the early stages of AD. In this study, transcriptome based on the Illumina RNA-seq and proteome based on TMT-labelling were performed for RNA and protein profiling on AD EC samples and non-AD control EC samples. Immunohistochemistry was used to validate proteins expressions. After integrated analysis, 57 genes were detected both in transcriptome and proteome data, including 51 in similar altering trends (7 upregulated, 44 downregulated) and 6 in inverse trends when compared AD vs. control. The top 6 genes (GABRG2, CACNG3, CACNB4, GABRB2, GRIK2, and SLC17A6) within the 51 genes were selected and related to "ion transport". Correlation analysis demonstrated negative relationship of protein expression level with the neuropathologic changes. In conclusion, the integrate transcriptome and proteome analysis provided evidence for dysregulation of ion transport across brain regions in AD, which might be a critical signaling pathway that initiates pathology. This study might provide new insight into the earliest changes occurring in the EC of AD and novel targets for AD prevention and treatment.
Collapse
Affiliation(s)
- Yangjie Jia
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Xia Wang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Yanyu Chen
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China.
| |
Collapse
|
17
|
Protein Phosphatase 2A (PP2A) mutations in brain function, development, and neurologic disease. Biochem Soc Trans 2021; 49:1567-1588. [PMID: 34241636 DOI: 10.1042/bst20201313] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022]
Abstract
By removing Ser/Thr-specific phosphorylations in a multitude of protein substrates in diverse tissues, Protein Phosphatase type 2A (PP2A) enzymes play essential regulatory roles in cellular signalling and physiology, including in brain function and development. Here, we review current knowledge on PP2A gene mutations causally involved in neurodevelopmental disorders and intellectual disability, focusing on PPP2CA, PPP2R1A and PPP2R5D. We provide insights into the impact of these mutations on PP2A structure, substrate specificity and potential function in neurobiology and brain development.
Collapse
|
18
|
Marcantoni A, Calorio C, Hidisoglu E, Chiantia G, Carbone E. Cav1.2 channelopathies causing autism: new hallmarks on Timothy syndrome. Pflugers Arch 2020; 472:775-789. [PMID: 32621084 DOI: 10.1007/s00424-020-02430-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Cav1.2 L-type calcium channels play key roles in long-term synaptic plasticity, sensory transduction, muscle contraction, and hormone release. De novo mutations in the gene encoding Cav1.2 (CACNA1C) causes two forms of Timothy syndrome (TS1, TS2), characterized by a multisystem disorder inclusive of cardiac arrhythmias, long QT, autism, and adrenal gland dysfunction. In both TS1 and TS2, the missense mutation G406R is on the alternatively spliced exon 8 and 8A coding for the IS6-helix of Cav1.2 and is responsible for the penetrant form of autism in most TS individuals. The mutation causes specific gain-of-function changes to Cav1.2 channel gating: a "leftward shift" of voltage-dependent activation, reduced voltage-dependent inactivation, and a "leftward shift" of steady-state inactivation. How this occurs and how Cav1.2 gating changes alter neuronal firing and synaptic plasticity is still largely unexplained. Trying to better understanding the molecular basis of Cav1.2 gating dysfunctions leading to autism, here, we will present and discuss the properties of recently reported typical and atypical TS phenotypes and the effective gating changes exhibited by missense mutations associated with long QTs without extracardiac symptoms, unrelated to TS. We will also discuss new emerging views achieved from using iPSCs-derived neurons and the newly available autistic TS2-neo mouse model, both appearing promising for understanding neuronal mistuning in autistic TS patients. We will also analyze and describe recent proposals of molecular pathways that might explain mistuned Ca2+-mediated and Ca2+-independent excitation-transcription signals to the nucleus. Briefly, we will also discuss possible pharmacological approaches to treat autism associated with L-type channelopathies.
Collapse
Affiliation(s)
- Andrea Marcantoni
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Chiara Calorio
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Enis Hidisoglu
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Giuseppe Chiantia
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Emilio Carbone
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy.
| |
Collapse
|
19
|
Coste de Bagneaux P, von Elsner L, Bierhals T, Campiglio M, Johannsen J, Obermair GJ, Hempel M, Flucher BE, Kutsche K. A homozygous missense variant in CACNB4 encoding the auxiliary calcium channel beta4 subunit causes a severe neurodevelopmental disorder and impairs channel and non-channel functions. PLoS Genet 2020; 16:e1008625. [PMID: 32176688 PMCID: PMC7176149 DOI: 10.1371/journal.pgen.1008625] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 04/22/2020] [Accepted: 01/23/2020] [Indexed: 12/27/2022] Open
Abstract
P/Q-type channels are the principal presynaptic calcium channels in brain functioning in neurotransmitter release. They are composed of the pore-forming CaV2.1 α1 subunit and the auxiliary α2δ-2 and β4 subunits. β4 is encoded by CACNB4, and its multiple splice variants serve isoform-specific functions as channel subunits and transcriptional regulators in the nucleus. In two siblings with intellectual disability, psychomotor retardation, blindness, epilepsy, movement disorder and cerebellar atrophy we identified rare homozygous variants in the genes LTBP1, EMILIN1, CACNB4, MINAR1, DHX38 and MYO15 by whole-exome sequencing. In silico tools, animal model, clinical, and genetic data suggest the p.(Leu126Pro) CACNB4 variant to be likely pathogenic. To investigate the functional consequences of the CACNB4 variant, we introduced the corresponding mutation L125P into rat β4b cDNA. Heterologously expressed wild-type β4b associated with GFP-CaV1.2 and accumulated in presynaptic boutons of cultured hippocampal neurons. In contrast, the β4b-L125P mutant failed to incorporate into calcium channel complexes and to cluster presynaptically. When co-expressed with CaV2.1 in tsA201 cells, β4b and β4b-L125P augmented the calcium current amplitudes, however, β4b-L125P failed to stably complex with α1 subunits. These results indicate that p.Leu125Pro disrupts the stable association of β4b with native calcium channel complexes, whereas membrane incorporation, modulation of current density and activation properties of heterologously expressed channels remained intact. Wildtype β4b was specifically targeted to the nuclei of quiescent excitatory cells. Importantly, the p.Leu125Pro mutation abolished nuclear targeting of β4b in cultured myotubes and hippocampal neurons. While binding of β4b to the known interaction partner PPP2R5D (B56δ) was not affected by the mutation, complex formation between β4b-L125P and the neuronal TRAF2 and NCK interacting kinase (TNIK) seemed to be disturbed. In summary, our data suggest that the homozygous CACNB4 p.(Leu126Pro) variant underlies the severe neurological phenotype in the two siblings, most likely by impairing both channel and non-channel functions of β4b.
Collapse
Affiliation(s)
| | - Leonie von Elsner
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marta Campiglio
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Jessika Johannsen
- Childrens Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerald J. Obermair
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
- Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard E. Flucher
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
20
|
Biswas D, Cary W, Nolta JA. PPP2R5D-Related Intellectual Disability and Neurodevelopmental Delay: A Review of the Current Understanding of the Genetics and Biochemical Basis of the Disorder. Int J Mol Sci 2020; 21:ijms21041286. [PMID: 32074998 PMCID: PMC7072873 DOI: 10.3390/ijms21041286] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
Protein Phosphatase 2 Regulatory Subunit B′ Delta (PPP2R5D)-related intellectual disability (ID) and neurodevelopmental delay results from germline de novo mutations in the PPP2R5D gene. This gene encodes the protein PPP2R5D (also known as the B56 delta subunit), which is an isoform of the subunit family B56 of the enzyme serine/threonine-protein phosphatase 2A (PP2A). Clinical signs include intellectual disability (ID); autism spectrum disorder (ASD); epilepsy; speech problems; behavioral challenges; and ophthalmologic, skeletal, endocrine, cardiac, and genital malformations. The association of defective PP2A activity in the brain with a wide range of severity of ID, along with its role in ASD, Alzheimer’s disease, and Parkinson’s-like symptoms, have recently generated the impetus for further research into mutations within this gene. PP2A, together with protein phosphatase 1 (PP1), accounts for more than 90% of all phospho-serine/threonine dephosphorylations in different tissues. The specificity for a wide variety of substrates is determined through nearly 100 different PP2A holoenzymes that are formed by at least 23 types of regulatory B subunits, and two isoforms each of the catalytic subunit C and the structural subunit A. In the mammalian brain, PP2A-mediated protein dephosphorylation plays an important role in learning and memory. The PPP2R5D subunit is highly expressed in the brain and the PPP2A–PPP2R5D holoenzyme plays an important role in maintaining neurons and regulating neuronal signaling. From 2015 to 2017, 25 individuals with PPP2R5D-related developmental disorder were diagnosed. Since then, Whole-Exome Sequencing (WES) has helped to identify more unrelated individuals clinically diagnosed with a neurodevelopmental disorder with pathological variants of PPP2R5D. In this review, we discuss the current understanding of the clinical and genetic aspects of the disorder in the context of the known functions of the PP2A–PPP2R5D holoenzyme in the brain, as well as the pathogenic mutations in PPP2R5D that lead to deficient PP2A–PPP2R5D dephosphorylation and their implications during development and in the etiology of autism, Parkinson’s disease, Alzheimer’s disease, and so forth. In the future, tools such as transgenic animals carrying pathogenic PPP2R5D mutations, and patient-derived induced pluripotent stem cell lines need to be developed in order to fully understand the effects of these mutations on different neural cell types.
Collapse
Affiliation(s)
- Dayita Biswas
- SPARK Program Scholar, Institute for Regenerative Cures, University of California, Sacramento, CA 95817, USA;
| | - Whitney Cary
- Stem Cell Program, UC Davis School of Medicine. The University of California, Sacramento, CA 95817, USA
- UC Davis Gene Therapy Program, University of California, Sacramento, CA 95817, USA
- Correspondence: (W.C.); (J.A.N.)
| | - Jan A. Nolta
- SPARK Program Scholar, Institute for Regenerative Cures, University of California, Sacramento, CA 95817, USA;
- Stem Cell Program, UC Davis School of Medicine. The University of California, Sacramento, CA 95817, USA
- UC Davis Gene Therapy Program, University of California, Sacramento, CA 95817, USA
- Correspondence: (W.C.); (J.A.N.)
| |
Collapse
|
21
|
Haworth AS, Brackenbury WJ. Emerging roles for multifunctional ion channel auxiliary subunits in cancer. Cell Calcium 2019; 80:125-140. [PMID: 31071485 PMCID: PMC6553682 DOI: 10.1016/j.ceca.2019.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023]
Abstract
Several superfamilies of plasma membrane channels which regulate transmembrane ion flux have also been shown to regulate a multitude of cellular processes, including proliferation and migration. Ion channels are typically multimeric complexes consisting of conducting subunits and auxiliary, non-conducting subunits. Auxiliary subunits modulate the function of conducting subunits and have putative non-conducting roles, further expanding the repertoire of cellular processes governed by ion channel complexes to processes such as transcellular adhesion and gene transcription. Given this expansive influence of ion channels on cellular behaviour it is perhaps no surprise that aberrant ion channel expression is a common occurrence in cancer. This review will focus on the conducting and non-conducting roles of the auxiliary subunits of various Ca2+, K+, Na+ and Cl- channels and the burgeoning evidence linking such auxiliary subunits to cancer. Several subunits are upregulated (e.g. Cavβ, Cavγ) and downregulated (e.g. Kvβ) in cancer, while other subunits have been functionally implicated as oncogenes (e.g. Navβ1, Cavα2δ1) and tumour suppressor genes (e.g. CLCA2, KCNE2, BKγ1) based on in vivo studies. The strengthening link between ion channel auxiliary subunits and cancer has exposed these subunits as potential biomarkers and therapeutic targets. However further mechanistic understanding is required into how these subunits contribute to tumour progression before their therapeutic potential can be fully realised.
Collapse
Affiliation(s)
- Alexander S Haworth
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK.
| |
Collapse
|
22
|
Guzman GA, Guzman RE, Jordan N, Hidalgo P. A Tripartite Interaction Among the Calcium Channel α 1- and β-Subunits and F-Actin Increases the Readily Releasable Pool of Vesicles and Its Recovery After Depletion. Front Cell Neurosci 2019; 13:125. [PMID: 31130843 PMCID: PMC6509170 DOI: 10.3389/fncel.2019.00125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/13/2019] [Indexed: 11/13/2022] Open
Abstract
Neurotransmitter release is initiated by the influx of Ca2+via voltage-gated calcium channels. The accessory β-subunit (CaVβ) of these channels shapes synaptic transmission by associating with the pore-forming subunit (CaVα1) and up-regulating presynaptic calcium currents. Besides CaVα1, CaVβ interacts with several partners including actin filaments (F-actin). These filaments are known to associate with synaptic vesicles (SVs) at the presynaptic terminals and support their translocation within different pools, but the role of CaVβ/F-actin association on synaptic transmission has not yet been explored. We here study how CaVβ4, the major calcium channel β isoform in mamalian brain, modifies synaptic transmission in concert with F-actin in cultured hippocampal neurons. We analyzed the effect of exogenous CaVβ4 before and after pharmacological disruption of the actin cytoskeleton and dissected calcium channel-dependent and -independent functions by comparing the effects of the wild-type subunit with the one bearing a double mutation that impairs binding to CaVα1. We found that exogenously expressed wild-type CaVβ4 enhances spontaneous and depolarization-evoked excitatory postsynaptic currents (EPSCs) without altering synaptogenesis. CaVβ4 increases the size of the readily releasable pool (RRP) of SVs at resting conditions and accelerates their recovery after depletion. The enhanced neurotransmitter release induced by CaVβ4 is abolished upon disruption of the actin cytoskeleton. The CaVα1 association-deficient CaVβ4 mutant associates with actin filaments, but neither alters postsynaptic responses nor the time course of the RRP recovery. Furthermore, this mutant protein preserves the ability to increase the RRP size. These results indicate that the interplay between CaVβ4 and F-actin also support the recruitment of SVs to the RRP in a CaVα1-independent manner. Our studies show an emerging role of CaVβ in determining SV maturation toward the priming state and its replenishment after release. We envision that this subunit plays a role in coupling exocytosis to endocytosis during the vesicle cycle.
Collapse
Affiliation(s)
- Gustavo A Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Raul E Guzman
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Nadine Jordan
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany
| | - Patricia Hidalgo
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, Jülich, Germany.,Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
23
|
Machol K, Rousseau J, Ehresmann S, Garcia T, Nguyen TTM, Spillmann RC, Sullivan JA, Shashi V, Jiang YH, Stong N, Fiala E, Willing M, Pfundt R, Kleefstra T, Cho MT, McLaughlin H, Rosello Piera M, Orellana C, Martínez F, Caro-Llopis A, Monfort S, Roscioli T, Nixon CY, Buckley MF, Turner A, Jones WD, van Hasselt PM, Hofstede FC, van Gassen KL, Brooks AS, van Slegtenhorst MA, Lachlan K, Sebastian J, Madan-Khetarpal S, Sonal D, Sakkubai N, Thevenon J, Faivre L, Maurel A, Petrovski S, Krantz ID, Tarpinian JM, Rosenfeld JA, Lee BH, Campeau PM, Adams DR, Alejandro ME, Allard P, Azamian MS, Bacino CA, Balasubramanyam A, Barseghyan H, Batzli GF, Beggs AH, Behnam B, Bican A, Bick DP, Birch CL, Bonner D, Boone BE, Bostwick BL, Briere LC, Brown DM, Brush M, Burke EA, Burrage LC, Chen S, Clark GD, Coakley TR, Cogan JD, Cooper CM, Cope H, Craigen WJ, D’Souza P, Davids M, Dayal JG, Dell’Angelica EC, Dhar SU, Dillon A, Dipple KM, Donnell-Fink LA, Dorrani N, Dorset DC, Douine ED, Draper DD, Eckstein DJ, Emrick LT, Eng CM, Eskin A, Esteves C, Estwick T, Ferreira C, Fogel BL, Friedman ND, Gahl WA, Glanton E, Godfrey RA, Goldstein DB, Gould SE, Gourdine JPF, et alMachol K, Rousseau J, Ehresmann S, Garcia T, Nguyen TTM, Spillmann RC, Sullivan JA, Shashi V, Jiang YH, Stong N, Fiala E, Willing M, Pfundt R, Kleefstra T, Cho MT, McLaughlin H, Rosello Piera M, Orellana C, Martínez F, Caro-Llopis A, Monfort S, Roscioli T, Nixon CY, Buckley MF, Turner A, Jones WD, van Hasselt PM, Hofstede FC, van Gassen KL, Brooks AS, van Slegtenhorst MA, Lachlan K, Sebastian J, Madan-Khetarpal S, Sonal D, Sakkubai N, Thevenon J, Faivre L, Maurel A, Petrovski S, Krantz ID, Tarpinian JM, Rosenfeld JA, Lee BH, Campeau PM, Adams DR, Alejandro ME, Allard P, Azamian MS, Bacino CA, Balasubramanyam A, Barseghyan H, Batzli GF, Beggs AH, Behnam B, Bican A, Bick DP, Birch CL, Bonner D, Boone BE, Bostwick BL, Briere LC, Brown DM, Brush M, Burke EA, Burrage LC, Chen S, Clark GD, Coakley TR, Cogan JD, Cooper CM, Cope H, Craigen WJ, D’Souza P, Davids M, Dayal JG, Dell’Angelica EC, Dhar SU, Dillon A, Dipple KM, Donnell-Fink LA, Dorrani N, Dorset DC, Douine ED, Draper DD, Eckstein DJ, Emrick LT, Eng CM, Eskin A, Esteves C, Estwick T, Ferreira C, Fogel BL, Friedman ND, Gahl WA, Glanton E, Godfrey RA, Goldstein DB, Gould SE, Gourdine JPF, Groden CA, Gropman AL, Haendel M, Hamid R, Hanchard NA, Handley LH, Herzog MR, Holm IA, Hom J, Howerton EM, Huang Y, Jacob HJ, Jain M, Jiang YH, Johnston JM, Jones AL, Kohane IS, Krasnewich DM, Krieg EL, Krier JB, Lalani SR, Lau CC, Lazar J, Lee BH, Lee H, Levy SE, Lewis RA, Lincoln SA, Lipson A, Loo SK, Loscalzo J, Maas RL, Macnamara EF, MacRae CA, Maduro VV, Majcherska MM, Malicdan MCV, Mamounas LA, Manolio TA, Markello TC, Marom R, Martínez-Agosto JA, Marwaha S, May T, McConkie-Rosell A, McCormack CE, McCray AT, Might M, Moretti PM, Morimoto M, Mulvihill JJ, Murphy JL, Muzny DM, Nehrebecky ME, Nelson SF, Newberry JS, Newman JH, Nicholas SK, Novacic D, Orange JS, Pallais JC, Palmer CG, Papp JC, Parker NH, Pena LD, Phillips JA, Posey JE, Postlethwait JH, Potocki L, Pusey BN, Reuter CM, Robertson AK, Rodan LH, Rosenfeld JA, Sampson JB, Samson SL, Schoch K, Schroeder MC, Scott DA, Sharma P, Shashi V, Signer R, Silverman EK, Sinsheimer JS, Smith KS, Spillmann RC, Splinter K, Stoler JM, Stong N, Sullivan JA, Sweetser DA, Tifft CJ, Toro C, Tran AA, Urv TK, Valivullah ZM, Vilain E, Vogel TP, Wahl CE, Walley NM, Walsh CA, Ward PA, Waters KM, Westerfield M, Wise AL, Wolfe LA, Worthey EA, Yamamoto S, Yang Y, Yu G, Zastrow DB, Zheng A. Expanding the Spectrum of BAF-Related Disorders: De Novo Variants in SMARCC2 Cause a Syndrome with Intellectual Disability and Developmental Delay. Am J Hum Genet 2019; 104:164-178. [PMID: 30580808 DOI: 10.1016/j.ajhg.2018.11.007] [Show More Authors] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
SMARCC2 (BAF170) is one of the invariable core subunits of the ATP-dependent chromatin remodeling BAF (BRG1-associated factor) complex and plays a crucial role in embryogenesis and corticogenesis. Pathogenic variants in genes encoding other components of the BAF complex have been associated with intellectual disability syndromes. Despite its significant biological role, variants in SMARCC2 have not been directly associated with human disease previously. Using whole-exome sequencing and a web-based gene-matching program, we identified 15 individuals with variable degrees of neurodevelopmental delay and growth retardation harboring one of 13 heterozygous variants in SMARCC2, most of them novel and proven de novo. The clinical presentation overlaps with intellectual disability syndromes associated with other BAF subunits, such as Coffin-Siris and Nicolaides-Baraitser syndromes and includes prominent speech impairment, hypotonia, feeding difficulties, behavioral abnormalities, and dysmorphic features such as hypertrichosis, thick eyebrows, thin upper lip vermilion, and upturned nose. Nine out of the fifteen individuals harbor variants in the highly conserved SMARCC2 DNA-interacting domains (SANT and SWIRM) and present with a more severe phenotype. Two of these individuals present cardiac abnormalities. Transcriptomic analysis of fibroblasts from affected individuals highlights a group of differentially expressed genes with possible roles in regulation of neuronal development and function, namely H19, SCRG1, RELN, and CACNB4. Our findings suggest a novel SMARCC2-related syndrome that overlaps with neurodevelopmental disorders associated with variants in BAF-complex subunits.
Collapse
|
24
|
Tammineni ER, Carrillo ED, Soto-Acosta R, Angel-Ambrocio AH, García MC, Bautista-Carbajal P, del Angel RM, Sánchez JA. The β
4
subunit of Ca
v
1.2 channels is required for an optimal interferon response in cardiac muscle cells. Sci Signal 2018; 11:11/560/eaaj1676. [DOI: 10.1126/scisignal.aaj1676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Eshwar R. Tammineni
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Elba D. Carrillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Rubén Soto-Acosta
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Antonio H. Angel-Ambrocio
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - María C. García
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Patricia Bautista-Carbajal
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Rosa M. del Angel
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Jorge A. Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| |
Collapse
|
25
|
β-Subunit of the voltage-gated Ca 2+ channel Cav1.2 drives signaling to the nucleus via H-Ras. Proc Natl Acad Sci U S A 2018; 115:E8624-E8633. [PMID: 30150369 DOI: 10.1073/pnas.1805380115] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Depolarization-induced signaling to the nucleus by the L-type voltage-gated calcium channel Cav1.2 is widely assumed to proceed by elevating intracellular calcium. The apparent lack of quantitative correlation between Ca2+ influx and gene activation suggests an alternative activation pathway. Here, we demonstrate that membrane depolarization of HEK293 cells transfected with α11.2/β2b/α2δ subunits (Cav1.2) triggers c-Fos and MeCP2 activation via the Ras/ERK/CREB pathway. Nuclear signaling is lost either by absence of the intracellular β2 subunit or by transfecting the cells with the channel mutant α11.2W440A/β2b/α2δ, a mutation that disrupts the interaction between α11.2 and β2 subunits. Pulldown assays in neuronal SH-SY5Y cells and in vitro binding of recombinant H-Ras and β2 confirmed the importance of the intracellular β2 subunit for depolarization-induced gene activation. Using a Ca2+-impermeable mutant channel α11.2L745P/β2b/α2δ or disrupting Ca2+/calmodulin binding to the channel using the channel mutant α11.2I1624A/β2b/α2δ, we demonstrate that depolarization-induced c-Fos and MeCP2 activation does not depend on Ca2+ transport by the channel. Thus, in contrast to the paradigm that elevated intracellular Ca2+ drives nuclear signaling, we show that Cav1.2-triggered c-Fos or MeCP2 is dependent on extracellular Ca2+ and Ca2+ occupancy of the open channel pore, but is Ca2+-influx independent. An indispensable β-subunit interaction with H-Ras, which is triggered by conformational changes at α11.2 independently of Ca2+ flux, brings to light a master regulatory role of β2 in transcriptional activation via the ERK/CREB pathway. This mode of H-Ras activation could have broad implications for understanding the coupling of membrane depolarization to the rapid induction of gene transcription.
Collapse
|
26
|
Lindy AS, Stosser MB, Butler E, Downtain‐Pickersgill C, Shanmugham A, Retterer K, Brandt T, Richard G, McKnight DA. Diagnostic outcomes for genetic testing of 70 genes in 8565 patients with epilepsy and neurodevelopmental disorders. Epilepsia 2018; 59:1062-1071. [DOI: 10.1111/epi.14074] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2018] [Indexed: 12/16/2022]
|
27
|
González-Ramírez R, Felix R. Transcriptional regulation of voltage-gated Ca 2+ channels. Acta Physiol (Oxf) 2018; 222. [PMID: 28371478 DOI: 10.1111/apha.12883] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/14/2017] [Accepted: 03/21/2017] [Indexed: 12/30/2022]
Abstract
The transcriptional regulation of voltage-gated Ca2+ (CaV ) channels is an emerging research area that promises to improve our understanding of how many relevant physiological events are shaped in the central nervous system, the skeletal muscle and other tissues. Interestingly, a picture of how transcription of CaV channel subunit genes is controlled is evolving with the identification of the promoter regions required for tissue-specific expression and the identification of transcription factors that control their expression. These promoters share several characteristics that include multiple transcriptional start sites, lack of a TATA box and the presence of elements conferring tissue-selective expression. Likewise, changes in CaV channel expression occur throughout development, following ischaemia, seizures or chronic drug administration. This review focuses on insights achieved regarding the control of CaV channel gene expression. To further understand the complexities of expression and to increase the possibilities of detecting CaV channel alterations causing human disease, a deeper knowledge on the structure of the 5' upstream regions of the genes encoding these remarkable proteins will be necessary.
Collapse
Affiliation(s)
- R. González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad; Hospital General ‘Dr. Manuel Gea González’; Secretaría de Salud; Ciudad de México México
| | - R. Felix
- Departmento de Biología Celular; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN); Ciudad de México México
| |
Collapse
|
28
|
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
|
29
|
Rima M, Daghsni M, Lopez A, Fajloun Z, Lefrancois L, Dunach M, Mori Y, Merle P, Brusés JL, De Waard M, Ronjat M. Down-regulation of the Wnt/β-catenin signaling pathway by Cacnb4. Mol Biol Cell 2017; 28:3699-3708. [PMID: 29021340 PMCID: PMC5706996 DOI: 10.1091/mbc.e17-01-0076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 11/29/2022] Open
Abstract
The β4 isoform of the β-subunits of voltage-gated calcium channel regulates cell proliferation and cell cycle progression. Herein we show that coexpression of the β4-subunit with actors of the canonical Wnt/β-catenin signaling pathway in a hepatoma cell line inhibits Wnt-responsive gene transcription and decreases cell division, in agreement with the role of the Wnt pathway in cell proliferation. β4-subunit-mediated inhibition of Wnt signaling is observed in the presence of LiCl, an inhibitor of glycogen synthase kinase (GSK3) that promotes β-catenin translocation to the nucleus. Expression of β4-subunit mutants that lost the ability to translocate to the nucleus has no effect on Wnt signaling, suggesting that β4-subunit inhibition of Wnt signaling occurs downstream from GSK3 and requires targeting of β4-subunit to the nucleus. β4-subunit coimmunoprecipitates with the TCF4 transcription factor and overexpression of TCF4 reverses the effect of β4-subunit on the Wnt pathway. We thus propose that the interaction of nuclear β4-subunit with TCF4 prevents β-catenin binding to TCF4 and leads to the inhibition of the Wnt-responsive gene transcription. Thereby, our results show that β4-subunit is a TCF4 repressor and therefore appears as an interesting candidate for the regulation of this pathway in neurons where β4-subunit is specifically expressed.
Collapse
Affiliation(s)
- Mohamad Rima
- L'institut du thorax, INSER, CNRS, Université de Nantes, 44000 Nantes, France
- LabEx Ion Channels Science and Therapeutics, Valbone 06560, France
- Azm Center for Research in Biotechnology and Its Application, Lebanese University, 1300 Tripoli, Lebanon
| | - Marwa Daghsni
- L'institut du thorax, INSER, CNRS, Université de Nantes, 44000 Nantes, France
- Université de Tunis El Manar, Faculté de Médecine de Tunis, LR99ES10 Laboratoire de Génétique Humaine, 1007 Tunis, Tunisie
| | - Anaïs Lopez
- INSERM U1052, Team on Hepatocarcinogenesis and Viral Infections, Centre of Research in Cancerology of Lyon, Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
- Hepatology Unit, Croix-Rousse Hospital, 69000 Lyon, France
| | - Ziad Fajloun
- Azm Center for Research in Biotechnology and Its Application, Lebanese University, 1300 Tripoli, Lebanon
| | - Lydie Lefrancois
- INSERM U1052, Team on Hepatocarcinogenesis and Viral Infections, Centre of Research in Cancerology of Lyon, Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
- Hepatology Unit, Croix-Rousse Hospital, 69000 Lyon, France
| | - Mireia Dunach
- Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Laboratory of Molecular Biology, Graduate School of Engineering, Kyoto University, 615-8510 Kyoto, Japan
| | - Philippe Merle
- INSERM U1052, Team on Hepatocarcinogenesis and Viral Infections, Centre of Research in Cancerology of Lyon, Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
- Hepatology Unit, Croix-Rousse Hospital, 69000 Lyon, France
| | - Juan L Brusés
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY 10522
| | - Michel De Waard
- L'institut du thorax, INSER, CNRS, Université de Nantes, 44000 Nantes, France
- LabEx Ion Channels Science and Therapeutics, Valbone 06560, France
- Smartox Biotechnology, 38400 Saint-Martin d'Hères, France
| | - Michel Ronjat
- L'institut du thorax, INSER, CNRS, Université de Nantes, 44000 Nantes, France
- LabEx Ion Channels Science and Therapeutics, Valbone 06560, France
| |
Collapse
|
30
|
Findeisen F, Campiglio M, Jo H, Abderemane-Ali F, Rumpf CH, Pope L, Rossen ND, Flucher BE, DeGrado WF, Minor DL. Stapled Voltage-Gated Calcium Channel (Ca V) α-Interaction Domain (AID) Peptides Act As Selective Protein-Protein Interaction Inhibitors of Ca V Function. ACS Chem Neurosci 2017; 8:1313-1326. [PMID: 28278376 PMCID: PMC5481814 DOI: 10.1021/acschemneuro.6b00454] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
![]()
For many voltage-gated
ion channels (VGICs), creation of a properly functioning ion channel
requires the formation of specific protein–protein interactions
between the transmembrane pore-forming subunits and cystoplasmic accessory
subunits. Despite the importance of such protein–protein interactions
in VGIC function and assembly, their potential as sites for VGIC modulator
development has been largely overlooked. Here, we develop meta-xylyl (m-xylyl) stapled peptides that
target a prototypic VGIC high affinity protein–protein interaction,
the interaction between the voltage-gated calcium channel (CaV) pore-forming subunit α-interaction domain (AID) and
cytoplasmic β-subunit (CaVβ). We show using
circular dichroism spectroscopy, X-ray crystallography, and isothermal
titration calorimetry that the m-xylyl staples enhance
AID helix formation are structurally compatible with native-like AID:CaVβ interactions and reduce the entropic penalty associated
with AID binding to CaVβ. Importantly, electrophysiological
studies reveal that stapled AID peptides act as effective inhibitors
of the CaVα1:CaVβ interaction
that modulate CaV function in an CaVβ
isoform-selective manner. Together, our studies provide a proof-of-concept
demonstration of the use of protein–protein interaction inhibitors
to control VGIC function and point to strategies for improved AID-based
CaV modulator design.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Daniel L. Minor
- Molecular Biophysics & Integrated Imaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
31
|
Rima M, Daghsni M, De Waard S, Gaborit N, Fajloun Z, Ronjat M, Mori Y, Brusés JL, De Waard M. The β 4 subunit of the voltage-gated calcium channel (Cacnb4) regulates the rate of cell proliferation in Chinese Hamster Ovary cells. Int J Biochem Cell Biol 2017; 89:57-70. [PMID: 28587927 DOI: 10.1016/j.biocel.2017.05.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/20/2017] [Accepted: 05/30/2017] [Indexed: 01/03/2023]
Abstract
The β subunits of Voltage-Gated Calcium Channel (VGCC) are cytosolic proteins that interact with the VGCC pore -forming subunit and participate in the trafficking of the channel to the cell membrane and in ion influx regulation. β subunits also exert functions independently of their binding to VGCC by translocation to the cell nucleus including the control of gene expression. Mutations of the neuronal Cacnb4 (β4) subunit are linked to human neuropsychiatric disorders including epilepsy and intellectual disabilities. It is believed that the pathogenic phenotype induced by these mutations is associated with channel-independent functions of the β4 subunit. In this report, we investigated the role of β4 subunit in cell proliferation and cell cycle progression and examined whether these functions could be altered by a pathogenic mutation. To this end, stably transfected Chinese Hamster Ovary (CHO-K1) cells expressing either rat full-length β4 or the rat C-terminally truncated epileptic mutant variant β1-481 were generated. The subcellular localization of both proteins differed significantly. Full-length β4 localizes almost exclusively in the cell nucleus and concentrates into the nucleolar compartment, while the C-terminal-truncated β1-481 subunit was less concentrated within the nucleus and absent from the nucleoli. Cell proliferation was found to be reduced by the expression of β4, while it was unaffected by the epileptic mutant. Also, full-length β4 interfered with cell cycle progression by presumably preventing cells from entering the S-phase via a mechanism that partially involves endogenous B56δ, a regulatory subunit of the phosphatase 2A (PP2A) that binds β4 but not β1-481. Analysis of β4 subcellular distribution during the cell cycle revealed that the protein is highly expressed in the nucleus at the G1/S transition phase and that it is translocated out of the nucleus during chromatin condensation and cell division. These results suggest that nuclear accumulation of β4 at the G1/S transition phase affects the progression into S-phase resulting in a decrease in the rate of cell proliferation. Regulation of the cell cycle exit is a critical step in determining the number of neuronal precursors and neuronal differentiation suggesting that mutations of the β4 subunit could affect neural development and formation of the mature central nervous system.
Collapse
Affiliation(s)
- Mohamad Rima
- INSERM UMR1087, LabEx Ion Channels Science and Therapeutics, Institut du Thorax, Nantes, F-44000 France; CNRS, UMR6291, Nantes, F-44000 France; Azm Center for Research in Biotechnology and its Application, Lebanese University, 1300, Tripoli, Lebanon
| | - Marwa Daghsni
- INSERM UMR1087, LabEx Ion Channels Science and Therapeutics, Institut du Thorax, Nantes, F-44000 France; CNRS, UMR6291, Nantes, F-44000 France; Université de Tunis El Manar, Faculté de Médecine de Tunis, LR99ES10 Laboratoire de Génétique Humaine, 1007, Tunis, Tunisia
| | - Stephan De Waard
- INSERM UMR1087, LabEx Ion Channels Science and Therapeutics, Institut du Thorax, Nantes, F-44000 France; CNRS, UMR6291, Nantes, F-44000 France; Université de Nantes, Nantes, F-44000 France
| | - Nathalie Gaborit
- INSERM UMR1087, LabEx Ion Channels Science and Therapeutics, Institut du Thorax, Nantes, F-44000 France; CNRS, UMR6291, Nantes, F-44000 France; Université de Nantes, Nantes, F-44000 France
| | - Ziad Fajloun
- Azm Center for Research in Biotechnology and its Application, Lebanese University, 1300, Tripoli, Lebanon; Faculty of Sciences III, Lebanese University, 1300, Tripoli, Lebanon
| | - Michel Ronjat
- INSERM UMR1087, LabEx Ion Channels Science and Therapeutics, Institut du Thorax, Nantes, F-44000 France; CNRS, UMR6291, Nantes, F-44000 France; Université de Nantes, Nantes, F-44000 France
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Juan L Brusés
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY 10522, USA
| | - Michel De Waard
- INSERM UMR1087, LabEx Ion Channels Science and Therapeutics, Institut du Thorax, Nantes, F-44000 France; CNRS, UMR6291, Nantes, F-44000 France; Université de Nantes, Nantes, F-44000 France.
| |
Collapse
|
32
|
Chu H, Sun P, Yin J, Liu G, Wang Y, Zhao P, Zhu Y, Yang X, Zheng T, Zhou X, Jin W, Sun C. Integrated network analysis reveals potentially novel molecular mechanisms and therapeutic targets of refractory epilepsies. PLoS One 2017; 12:e0174964. [PMID: 28388656 PMCID: PMC5384674 DOI: 10.1371/journal.pone.0174964] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 03/19/2017] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a complex neurological disorder and a significant health problem. The pathogenesis of epilepsy remains obscure in a significant number of patients and the current treatment options are not adequate in about a third of individuals which were known as refractory epilepsies (RE). Network medicine provides an effective approach for studying the molecular mechanisms underlying complex diseases. Here we integrated 1876 disease-gene associations of RE and located those genes to human protein-protein interaction (PPI) network to obtain 42 significant RE-associated disease modules. The functional analysis of these disease modules showed novel molecular pathological mechanisms of RE, such as the novel enriched pathways (e.g., "presynaptic nicotinic acetylcholine receptors", "signaling by insulin receptor"). Further analysis on the relationships between current drug targets and the RE-related disease genes showed the rational mechanisms of most antiepileptic drugs. In addition, we detected ten potential novel drug targets (e.g., KCNA1, KCNA4-6, KCNC3, KCND2, KCNMA1, CAMK2G, CACNB4 and GRM1) located in three RE related disease modules, which might provide novel insights into the new drug discovery for RE therapy.
Collapse
Affiliation(s)
- Hongwei Chu
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian, China
| | - Pin Sun
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiahui Yin
- College of Electronics and Information Engineering, Tongji University, Shanghai, China
| | - Guangming Liu
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Yiwei Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian, China
| | - Pengyao Zhao
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Yizhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaohan Yang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian, China
| | - Tiezheng Zheng
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian, China
| | - Xuezhong Zhou
- School of Computer and Information Technology and Beijing Key Lab of Traffic Data Analysis and Mining, Beijing Jiaotong University, Beijing, China
| | - Weilin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Changkai Sun
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Institute for Brain Disorders, Dalian Medical University, Dalian, China
- Research Center for the Control Engineering of Translational Precision Medicine, Dalian University of Technology, Dalian, China
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
33
|
Faridi U, Dhawan SS, Pal S, Gupta S, Shukla AK, Darokar MP, Sharma A, Shasany AK. Repurposing L-Menthol for Systems Medicine and Cancer Therapeutics? L-Menthol Induces Apoptosis through Caspase 10 and by Suppressing HSP90. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:53-64. [PMID: 26760959 DOI: 10.1089/omi.2015.0118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The objective of the present study was to repurpose L-menthol, which is frequently used in oral health and topical formulations, for cancer therapeutics. In this article, we argue that monoterpenes such as L-menthol might offer veritable potentials in systems medicine, for example, as cheaper anti-cancer compounds. Other monoterpenes such as limonene, perillyl alcohol, and geraniol have been shown to induce apoptosis in various cancer cell lines, but their mechanisms of action are yet to be completely elucidated. Earlier, we showed that L-menthol modulates tubulin polymerization and apoptosis to inhibit cancer cell proliferation. In the present report, we used an apoptosis-related gene microarray in conjunction with proteomics analyses, as well as in silico interpretations, to study gene expression modulation in human adenocarcinoma Caco-2 cell line in response to L-menthol treatment. The microarray analysis identified caspase 10 as the important initiator caspase, instead of caspase 8. The proteomics analyses showed downregulation of HSP90 protein (also corroborated by its low transcript abundance), which in turn indicated inhibition of AKT-mediated survival pathway, release of pro-apoptotic factor BAD from BAD and BCLxL complex, besides regulation of other factors related to apoptosis. Based on the combined microarray, proteomics, and in silico data, a signaling pathway for L-menthol-induced apoptosis is being presented for the first time here. These data and literature analysis have significant implications for "repurposing" L-menthol beyond oral medicine, and in understanding the mode of action of plant-derived monoterpenes towards development of cheaper anticancer drugs in future.
Collapse
Affiliation(s)
- Uzma Faridi
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Sunita S Dhawan
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Shaifali Pal
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Sanchita Gupta
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ashutosh K Shukla
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Mahendra P Darokar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ajit K Shasany
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| |
Collapse
|
34
|
Protein partners of the calcium channel β subunit highlight new cellular functions. Biochem J 2016; 473:1831-44. [DOI: 10.1042/bcj20160125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/15/2016] [Indexed: 12/21/2022]
Abstract
Calcium plays a key role in cell signalling by its intervention in a wide range of physiological processes. Its entry into cells occurs mainly via voltage-gated calcium channels (VGCC), which are found not only in the plasma membrane of excitable cells but also in cells insensitive to electrical signals. VGCC are composed of different subunits, α1, β, α2δ and γ, among which the cytosolic β subunit (Cavβ) controls the trafficking of the channel to the plasma membrane, its regulation and its gating properties. For many years, these were the main functions associated with Cavβ. However, a growing number of proteins have been found to interact with Cavβ, emphasizing the multifunctional role of this versatile protein. Interestingly, some of the newly assigned functions of Cavβ are independent of its role in the regulation of VGCC, and thus further increase its functional roles. Based on the identity of Cavβ protein partners, this review emphasizes the diverse cellular functions of Cavβ and summarizes both past findings as well as recent progress in the understanding of VGCC.
Collapse
|
35
|
Benedetti B, Benedetti A, Flucher BE. Loss of the calcium channel β4 subunit impairs parallel fibre volley and Purkinje cell firing in cerebellum of adult ataxic mice. Eur J Neurosci 2016; 43:1486-98. [PMID: 27003325 PMCID: PMC4949674 DOI: 10.1111/ejn.13241] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 01/08/2023]
Abstract
The auxiliary voltage-gated calcium channel subunit β4 supports targeting of calcium channels to the cell membrane, modulates ionic currents and promotes synaptic release in the central nervous system. β4 is abundant in cerebellum and its loss causes ataxia. However, the type of calcium channels and cerebellar functions affected by the loss of β4 are currently unknown. We therefore studied the structure and function of Purkinje cells in acute cerebellar slices of the β4 (-/-) ataxic (lethargic) mouse, finding that loss of β4 affected Purkinje cell input, morphology and pacemaker activity. In adult lethargic cerebellum evoked postsynaptic currents from parallel fibres were depressed, while paired-pulse facilitation and spontaneous synaptic currents were unaffected. Because climbing fibre input was spared, the parallel fibre/climbing fibre input ratio was reduced. The dendritic arbor of adult lethargic Purkinje cells displayed fewer and shorter dendrites, but a normal spine density. Accordingly, the width of the molecular and granular layers was reduced. These defects recapitulate the impaired cerebellar maturation observed upon Cav 2.1 ataxic mutations. However, unlike Cav 2.1 mutations, lethargic Purkinje cells also displayed a striking decrease in pacemaker firing frequency, without loss of firing regularity. All these deficiencies appear in late development, indicating the importance of β4 for the normal differentiation and function of mature Purkinje cells networks. The observed reduction of the parallel fibre input, the altered parallel fibre/climbing fibre ratio and the reduced Purkinje cell output can contribute to the severe motor impairment caused by the loss of the calcium channel β4 subunit in lethargic mice.
Collapse
Affiliation(s)
- Bruno Benedetti
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Schöpfstraße 41, Innsbruck, A-6020, Austria
| | - Ariane Benedetti
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Schöpfstraße 41, Innsbruck, A-6020, Austria
| | - Bernhard E Flucher
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Schöpfstraße 41, Innsbruck, A-6020, Austria
| |
Collapse
|
36
|
Abstract
According to the standard model of G protein-coupled receptor (GPCR) signaling, GPCRs are localized to the cell membrane where they respond to extracellular signals. Stimulation of GPCRs leads to the activation of heterotrimeric G proteins and their intracellular signaling pathways. However, this model fails to accommodate GPCRs, G proteins, and their downstream effectors that are found on the nuclear membrane or in the nucleus. Evidence from isolated nuclei indicates the presence of GPCRs on the nuclear membrane that can activate similar G protein-dependent signaling pathways in the nucleus as at the cell surface. These pathways also include activation of cyclic adenosine monophosphate, calcium and nitric oxide synthase signaling in cardiomyocytes. In addition, a number of distinct heterotrimeric and monomeric G proteins have been found in the nucleus of various cell types. This review will focus on understanding the function of nuclear G proteins with a focus on cardiac signaling where applicable.
Collapse
|
37
|
Shang L, Henderson LB, Cho MT, Petrey DS, Fong CT, Haude KM, Shur N, Lundberg J, Hauser N, Carmichael J, Innis J, Schuette J, Wu YW, Asaikar S, Pearson M, Folk L, Retterer K, Monaghan KG, Chung WK. De novo missense variants in PPP2R5D are associated with intellectual disability, macrocephaly, hypotonia, and autism. Neurogenetics 2015; 17:43-9. [PMID: 26576547 DOI: 10.1007/s10048-015-0466-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/22/2015] [Indexed: 12/19/2022]
Abstract
Protein phosphatase 2A (PP2A) is a heterotrimeric protein serine/threonine phosphatase and is involved in a broad range of cellular processes. PPP2R5D is a regulatory B subunit of PP2A and plays an important role in regulating key neuronal and developmental regulation processes such as PI3K/AKT and glycogen synthase kinase 3 beta (GSK3β)-mediated cell growth, chromatin remodeling, and gene transcriptional regulation. Using whole-exome sequencing (WES), we identified four de novo variants in PPP2R5D in a total of seven unrelated individuals with intellectual disability (ID) and other shared clinical characteristics, including autism spectrum disorder, macrocephaly, hypotonia, seizures, and dysmorphic features. Among the four variants, two have been previously reported and two are novel. All four amino acids are highly conserved among the PP2A subunit family, and all change a negatively charged acidic glutamic acid (E) to a positively charged basic lysine (K) and are predicted to disrupt the PP2A subunit binding and impair the dephosphorylation capacity. Our data provides further support for PPP2R5D as a genetic cause of ID.
Collapse
Affiliation(s)
- Linshan Shang
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | | | | | - Donald S Petrey
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA
| | - Chin-To Fong
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | | | | | - Jeffrey Innis
- Division of Pediatric Genetics, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jane Schuette
- Division of Pediatric Genetics, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yvonne W Wu
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
38
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 786] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
39
|
Campiglio M, Flucher BE. The role of auxiliary subunits for the functional diversity of voltage-gated calcium channels. J Cell Physiol 2015; 230:2019-31. [PMID: 25820299 PMCID: PMC4672716 DOI: 10.1002/jcp.24998] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 03/23/2015] [Indexed: 11/18/2022]
Abstract
Voltage-gated calcium channels (VGCCs) represent the sole mechanism to convert membrane depolarization into cellular functions like secretion, contraction, or gene regulation. VGCCs consist of a pore-forming α(1) subunit and several auxiliary channel subunits. These subunits come in multiple isoforms and splice-variants giving rise to a stunning molecular diversity of possible subunit combinations. It is generally believed that specific auxiliary subunits differentially regulate the channels and thereby contribute to the great functional diversity of VGCCs. If auxiliary subunits can associate and dissociate from pre-existing channel complexes, this would allow dynamic regulation of channel properties. However, most auxiliary subunits modulate current properties very similarly, and proof that any cellular calcium channel function is indeed modulated by the physiological exchange of auxiliary subunits is still lacking. In this review we summarize available information supporting a differential modulation of calcium channel functions by exchange of auxiliary subunits, as well as experimental evidence in support of alternative functions of the auxiliary subunits. At the heart of the discussion is the concept that, in their native environment, VGCCs function in the context of macromolecular signaling complexes and that the auxiliary subunits help to orchestrate the diverse protein-protein interactions found in these calcium channel signalosomes. Thus, in addition to a putative differential modulation of current properties, differential subcellular targeting properties and differential protein-protein interactions of the auxiliary subunits may explain the need for their vast molecular diversity.
Collapse
Affiliation(s)
- Marta Campiglio
- Division of Physiology, Department of Physiology and Medical Physics, Medical University InnsbruckInnsbruck, Austria
| | - Bernhard E Flucher
- Division of Physiology, Department of Physiology and Medical Physics, Medical University InnsbruckInnsbruck, Austria
| |
Collapse
|
40
|
Houge G, Haesen D, Vissers LELM, Mehta S, Parker MJ, Wright M, Vogt J, McKee S, Tolmie JL, Cordeiro N, Kleefstra T, Willemsen MH, Reijnders MRF, Berland S, Hayman E, Lahat E, Brilstra EH, van Gassen KLI, Zonneveld-Huijssoon E, de Bie CI, Hoischen A, Eichler EE, Holdhus R, Steen VM, Døskeland SO, Hurles ME, FitzPatrick DR, Janssens V. B56δ-related protein phosphatase 2A dysfunction identified in patients with intellectual disability. J Clin Invest 2015; 125:3051-62. [PMID: 26168268 DOI: 10.1172/jci79860] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 05/27/2015] [Indexed: 12/17/2022] Open
Abstract
Here we report inherited dysregulation of protein phosphatase activity as a cause of intellectual disability (ID). De novo missense mutations in 2 subunits of serine/threonine (Ser/Thr) protein phosphatase 2A (PP2A) were identified in 16 individuals with mild to severe ID, long-lasting hypotonia, epileptic susceptibility, frontal bossing, mild hypertelorism, and downslanting palpebral fissures. PP2A comprises catalytic (C), scaffolding (A), and regulatory (B) subunits that determine subcellular anchoring, substrate specificity, and physiological function. Ten patients had mutations within a highly conserved acidic loop of the PPP2R5D-encoded B56δ regulatory subunit, with the same E198K mutation present in 6 individuals. Five patients had mutations in the PPP2R1A-encoded scaffolding Aα subunit, with the same R182W mutation in 3 individuals. Some Aα cases presented with large ventricles, causing macrocephaly and hydrocephalus suspicion, and all cases exhibited partial or complete corpus callosum agenesis. Functional evaluation revealed that mutant A and B subunits were stable and uncoupled from phosphatase activity. Mutant B56δ was A and C binding-deficient, while mutant Aα subunits bound B56δ well but were unable to bind C or bound a catalytically impaired C, suggesting a dominant-negative effect where mutant subunits hinder dephosphorylation of B56δ-anchored substrates. Moreover, mutant subunit overexpression resulted in hyperphosphorylation of GSK3β, a B56δ-regulated substrate. This effect was in line with clinical observations, supporting a correlation between the ID degree and biochemical disturbance.
Collapse
|
41
|
Impact of gold nanoparticles on brain of mice infected with Schistosoma mansoni. Parasitol Res 2015; 114:3711-9. [DOI: 10.1007/s00436-015-4600-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 12/22/2022]
|
42
|
Etemad S, Campiglio M, Obermair GJ, Flucher BE. The juvenile myoclonic epilepsy mutant of the calcium channel β(4) subunit displays normal nuclear targeting in nerve and muscle cells. Channels (Austin) 2015; 8:334-43. [PMID: 24875574 PMCID: PMC4203735 DOI: 10.4161/chan.29322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Voltage-gated calcium channels regulate gene expression by controlling calcium entry through the plasma membrane and by direct interactions of channel fragments and auxiliary β subunits with promoters and the epigenetic machinery in the nucleus. Mutations of the calcium channel β4 subunit gene (CACNB4) cause juvenile myoclonic epilepsy in humans and ataxia and epileptic seizures in mice. Recently a model has been proposed according to which failed nuclear translocation of the truncated β4 subunit R482X mutation resulted in altered transcriptional regulation and consequently in neurological disease. Here we examined the nuclear targeting properties of the truncated β4b(1–481) subunit in tsA-201 cells, skeletal myotubes, and in hippocampal neurons. Contrary to expectation, nuclear targeting of β4b(1–481) was not reduced compared with full-length β4b in any one of the three cell systems. These findings oppose an essential role of the β4 distal C-terminus in nuclear targeting and challenge the idea that the nuclear function of calcium channel β4 subunits is critically involved in the etiology of epilepsy and ataxia in patients and mouse models with mutations in the CACNB4 gene.
Collapse
|
43
|
Nakao A, Miki T, Shoji H, Nishi M, Takeshima H, Miyakawa T, Mori Y. Comprehensive behavioral analysis of voltage-gated calcium channel beta-anchoring and -regulatory protein knockout mice. Front Behav Neurosci 2015; 9:141. [PMID: 26136667 PMCID: PMC4468383 DOI: 10.3389/fnbeh.2015.00141] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/13/2015] [Indexed: 01/20/2023] Open
Abstract
Calcium (Ca2+) influx through voltage-gated Ca2+ channels (VGCCs) induces numerous intracellular events such as neuronal excitability, neurotransmitter release, synaptic plasticity, and gene regulation. It has been shown that genes related to Ca2+ signaling, such as the CACNA1C, CACNB2, and CACNA1I genes that encode VGCC subunits, are associated with schizophrenia and other psychiatric disorders. Recently, VGCC beta-anchoring and -regulatory protein (BARP) was identified as a novel regulator of VGCC activity via the interaction of VGCC β subunits. To examine the role of the BARP in higher brain functions, we generated BARP knockout (KO) mice and conducted a comprehensive battery of behavioral tests. BARP KO mice exhibited greatly reduced locomotor activity, as evidenced by decreased vertical activity, stereotypic counts in the open field test, and activity level in the home cage, and longer latency to complete a session in spontaneous T-maze alteration test, which reached “study-wide significance.” Acoustic startle response was also reduced in the mutants. Interestingly, they showed multiple behavioral phenotypes that are seemingly opposite to those seen in the mouse models of schizophrenia and its related disorders, including increased working memory, flexibility, prepulse inhibition, and social interaction, and decreased locomotor activity, though many of these phenotypes are statistically weak and require further replications. These results demonstrate that BARP is involved in the regulation of locomotor activity and, possibly, emotionality. The possibility was also suggested that BARP KO mice may serve as a unique tool for investigating the pathogenesis/pathophysiology of schizophrenia and related disorders. Further evaluation of the molecular and physiological phenotypes of the mutant mice would provide new insights into the role of BARP in higher brain functions.
Collapse
Affiliation(s)
- Akito Nakao
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University Toyoake, Japan
| | - Takafumi Miki
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University Toyoake, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Kawaguchi, Japan
| | - Miyuki Nishi
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University Kyoto, Japan
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University Kyoto, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University Toyoake, Japan ; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology Kawaguchi, Japan ; Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences Okazaki, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| |
Collapse
|
44
|
Gstir R, Schafferer S, Scheideler M, Misslinger M, Griehl M, Daschil N, Humpel C, Obermair GJ, Schmuckermair C, Striessnig J, Flucher BE, Hüttenhofer A. Generation of a neuro-specific microarray reveals novel differentially expressed noncoding RNAs in mouse models for neurodegenerative diseases. RNA (NEW YORK, N.Y.) 2014; 20:1929-43. [PMID: 25344396 PMCID: PMC4238357 DOI: 10.1261/rna.047225.114] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/27/2014] [Indexed: 05/24/2023]
Abstract
We have generated a novel, neuro-specific ncRNA microarray, covering 1472 ncRNA species, to investigate their expression in different mouse models for central nervous system diseases. Thereby, we analyzed ncRNA expression in two mouse models with impaired calcium channel activity, implicated in Epilepsy or Parkinson's disease, respectively, as well as in a mouse model mimicking pathophysiological aspects of Alzheimer's disease. We identified well over a hundred differentially expressed ncRNAs, either from known classes of ncRNAs, such as miRNAs or snoRNAs or which represented entirely novel ncRNA species. Several differentially expressed ncRNAs in the calcium channel mouse models were assigned as miRNAs and target genes involved in calcium signaling, thus suggesting feedback regulation of miRNAs by calcium signaling. In the Alzheimer mouse model, we identified two snoRNAs, whose expression was deregulated prior to amyloid plaque formation. Interestingly, the presence of snoRNAs could be detected in cerebral spine fluid samples in humans, thus potentially serving as early diagnostic markers for Alzheimer's disease. In addition to known ncRNAs species, we also identified 63 differentially expressed, entirely novel ncRNA candidates, located in intronic or intergenic regions of the mouse genome, genomic locations, which previously have been shown to harbor the majority of functional ncRNAs.
Collapse
Affiliation(s)
- Ronald Gstir
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Simon Schafferer
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Marcel Scheideler
- RNA Biology Group, Institute for Genomics and Bioinformatics, Graz University of Technology, 8010 Graz, Austria
| | - Matthias Misslinger
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Matthias Griehl
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Nina Daschil
- Department of Psychiatry and Psychotherapy, University Clinic of General and Social Psychiatry, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Christian Humpel
- Department of Psychiatry and Psychotherapy, University Clinic of General and Social Psychiatry, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Claudia Schmuckermair
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Joerg Striessnig
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Bernhard E Flucher
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Alexander Hüttenhofer
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
45
|
Profiling status epilepticus-induced changes in hippocampal RNA expression using high-throughput RNA sequencing. Sci Rep 2014; 4:6930. [PMID: 25373493 PMCID: PMC4894418 DOI: 10.1038/srep06930] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/09/2014] [Indexed: 12/30/2022] Open
Abstract
Status epilepticus (SE) is a life-threatening condition that can give rise to a number of neurological disorders, including learning deficits, depression, and epilepsy. Many of the effects of SE appear to be mediated by alterations in gene expression. To gain deeper insight into how SE affects the transcriptome, we employed the pilocarpine SE model in mice and Illumina-based high-throughput sequencing to characterize alterations in gene expression from the induction of SE, to the development of spontaneous seizure activity. While some genes were upregulated over the entire course of the pathological progression, each of the three sequenced time points (12-hour, 10-days and 6-weeks post-SE) had a largely unique transcriptional profile. Hence, genes that regulate synaptic physiology and transcription were most prominently altered at 12-hours post-SE; at 10-days post-SE, marked changes in metabolic and homeostatic gene expression were detected; at 6-weeks, substantial changes in the expression of cell excitability and morphogenesis genes were detected. At the level of cell signaling, KEGG analysis revealed dynamic changes within the MAPK pathways, as well as in CREB-associated gene expression. Notably, the inducible expression of several noncoding transcripts was also detected. These findings offer potential new insights into the cellular events that shape SE-evoked pathology.
Collapse
|
46
|
Taylor J, Pereyra A, Zhang T, Messi ML, Wang ZM, Hereñú C, Kuan PF, Delbono O. The Cavβ1a subunit regulates gene expression and suppresses myogenin in muscle progenitor cells. ACTA ACUST UNITED AC 2014; 205:829-46. [PMID: 24934157 PMCID: PMC4068134 DOI: 10.1083/jcb.201403021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cavβ1a acts as a voltage-gated calcium channel-independent regulator of gene expression in muscle progenitor cells and is required for their normal expansion during myogenic development. Voltage-gated calcium channel (Cav) β subunits are auxiliary subunits to Cavs. Recent reports show Cavβ subunits may enter the nucleus and suggest a role in transcriptional regulation, but the physiological relevance of this localization remains unclear. We sought to define the nuclear function of Cavβ in muscle progenitor cells (MPCs). We found that Cavβ1a is expressed in proliferating MPCs, before expression of the calcium conducting subunit Cav1.1, and enters the nucleus. Loss of Cavβ1a expression impaired MPC expansion in vitro and in vivo and caused widespread changes in global gene expression, including up-regulation of myogenin. Additionally, we found that Cavβ1a localizes to the promoter region of a number of genes, preferentially at noncanonical (NC) E-box sites. Cavβ1a binds to a region of the Myog promoter containing an NC E-box, suggesting a mechanism for inhibition of myogenin gene expression. This work indicates that Cavβ1a acts as a Cav-independent regulator of gene expression in MPCs, and is required for their normal expansion during myogenic development.
Collapse
Affiliation(s)
- Jackson Taylor
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Andrea Pereyra
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157 Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, 1900 La Plata, BA, Argentina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Claudia Hereñú
- Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, 1900 La Plata, BA, Argentina
| | - Pei-Fen Kuan
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
47
|
Simms BA, Zamponi GW. Neuronal voltage-gated calcium channels: structure, function, and dysfunction. Neuron 2014; 82:24-45. [PMID: 24698266 DOI: 10.1016/j.neuron.2014.03.016] [Citation(s) in RCA: 456] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Voltage-gated calcium channels are the primary mediators of depolarization-induced calcium entry into neurons. There is great diversity of calcium channel subtypes due to multiple genes that encode calcium channel α1 subunits, coassembly with a variety of ancillary calcium channel subunits, and alternative splicing. This allows these channels to fulfill highly specialized roles in specific neuronal subtypes and at particular subcellular loci. While calcium channels are of critical importance to brain function, their inappropriate expression or dysfunction gives rise to a variety of neurological disorders, including, pain, epilepsy, migraine, and ataxia. This Review discusses salient aspects of voltage-gated calcium channel function, physiology, and pathophysiology.
Collapse
Affiliation(s)
- Brett A Simms
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
48
|
Differential neuronal targeting of a new and two known calcium channel β4 subunit splice variants correlates with their regulation of gene expression. J Neurosci 2014; 34:1446-61. [PMID: 24453333 DOI: 10.1523/jneurosci.3935-13.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The β subunits of voltage-gated calcium channels regulate surface expression and gating of CaV1 and CaV2 α1 subunits and thus contribute to neuronal excitability, neurotransmitter release, and calcium-induced gene regulation. In addition, certain β subunits are targeted into the nucleus, where they interact directly with the epigenetic machinery. Whereas their involvement in this multitude of functions is reflected by a great molecular heterogeneity of β isoforms derived from four genes and abundant alternative splicing, little is known about the roles of individual β variants in specific neuronal functions. In the present study, an alternatively spliced β4 subunit lacking the variable N terminus (β4e) is identified. It is highly expressed in mouse cerebellum and cultured cerebellar granule cells (CGCs) and modulates P/Q-type calcium currents in tsA201 cells and CaV2.1 surface expression in neurons. Compared with the other two known full-length β4 variants (β4a and β4b), β4e is most abundantly expressed in the distal axon, but lacks nuclear-targeting properties. To determine the importance of nuclear targeting of β4 subunits for transcriptional regulation, we performed whole-genome expression profiling of CGCs from lethargic (β4-null) mice individually reconstituted with β4a, β4b, and β4e. Notably, the number of genes regulated by each β4 splice variant correlated with the rank order of their nuclear-targeting properties (β4b > β4a > β4e). Together, these findings support isoform-specific functions of β4 splice variants in neurons, with β4b playing a dual role in channel modulation and gene regulation, whereas the newly detected β4e variant serves exclusively in calcium-channel-dependent functions.
Collapse
|
49
|
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
|
50
|
Felix R, Calderón-Rivera A, Andrade A. Regulation of high-voltage-activated Ca 2+ channel function, trafficking, and membrane stability by auxiliary subunits. ACTA ACUST UNITED AC 2013; 2:207-220. [PMID: 24949251 DOI: 10.1002/wmts.93] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Voltage-gated Ca2+ (CaV) channels mediate Ca2+ ions influx into cells in response to depolarization of the plasma membrane. They are responsible for initiation of excitation-contraction and excitation-secretion coupling, and the Ca2+ that enters cells through this pathway is also important in the regulation of protein phosphorylation, gene transcription, and many other intracellular events. Initial electrophysiological studies divided CaV channels into low-voltage-activated (LVA) and high-voltage-activated (HVA) channels. The HVA CaV channels were further subdivided into L, N, P/Q, and R-types which are oligomeric protein complexes composed of an ion-conducting CaVα1 subunit and auxiliary CaVα2δ, CaVβ, and CaVγ subunits. The functional consequences of the auxiliary subunits include altered functional and pharmacological properties of the channels as well as increased current densities. The latter observation suggests an important role of the auxiliary subunits in membrane trafficking of the CaVα1 subunit. This includes the mechanisms by which CaV channels are targeted to the plasma membrane and to appropriate regions within a given cell. Likewise, the auxiliary subunits seem to participate in the mechanisms that remove CaV channels from the plasma membrane for recycling and/or degradation. Diverse studies have provided important clues to the molecular mechanisms involved in the regulation of CaV channels by the auxiliary subunits, and the roles that these proteins could possibly play in channel targeting and membrane Stabilization.
Collapse
Affiliation(s)
- Ricardo Felix
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| | - Aida Calderón-Rivera
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| | - Arturo Andrade
- Department of Neuroscience, Brown University, Providence, RI, USA
| |
Collapse
|