1
|
de la Rosa Vázquez J, Lee A. Role of the C-terminal domain in modifying pH-dependent regulation of Ca v1.4 Ca 2+ channels. Channels (Austin) 2025; 19:2473074. [PMID: 40116026 PMCID: PMC11934190 DOI: 10.1080/19336950.2025.2473074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/23/2025] Open
Abstract
In the retina, Ca2+ influx through Cav1.4 Ca2+ channels triggers neurotransmitter release from rod and cone photoreceptors. Changes in extracellular pH modify channel opening, enabling a feedback regulation of photoreceptor output that contributes to the encoding of color and contrast. However, the mechanisms underlying pH-dependent modulation of Cav1.4 are poorly understood. Here, we investigated the role of the C-terminal domain (CTD) of Cav1.4 in pH-dependent modulation of Ba2+ currents (IBa) in HEK293T cells transfected with the full length CaV1.4 (FL) or variants lacking portions of the CTD due to alternative splicing (Δe47) or a disease-causing mutation (K1591X). While extracellular alkalinization caused an increase in IBa for each variant, the magnitude of this increase was significantly diminished (~40-50%) for both CTD variants; K1591X was unique in showing no pH-dependent increase in maximal conductance. Moreover, the auxiliary α2δ-4 subunit augmented the pH sensitivity of IBa, as compared to α2δ-1 or no α2δ, for FL and K1591X but not Δe47. We conclude that the CTD and α2δ-4 are critical determinants of pH-dependent modulation of Cav1.4 and may influence the processing of visual information in normal and diseased states of the retina.
Collapse
Affiliation(s)
- Juan de la Rosa Vázquez
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX, USA
| | - Amy Lee
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
2
|
Limpitikul WB, Dick IE. Inactivation of CaV1 and CaV2 channels. J Gen Physiol 2025; 157:e202313531. [PMID: 39883005 PMCID: PMC11781272 DOI: 10.1085/jgp.202313531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) are highly expressed throughout numerous biological systems and play critical roles in synaptic transmission, cardiac excitation, and muscle contraction. To perform these various functions, VGCCs are highly regulated. Inactivation comprises a critical mechanism controlling the entry of Ca2+ through these channels and constitutes an important means to regulate cellular excitability, shape action potentials, control intracellular Ca2+ levels, and contribute to long-term potentiation and depression. For CaV1 and CaV2 channel families, inactivation proceeds via two distinct processes. Voltage-dependent inactivation (VDI) reduces Ca2+ entry through the channel in response to sustained or repetitive depolarization, while Ca2+-dependent inactivation (CDI) occurs in response to elevations in intracellular Ca2+ levels. These processes are critical for physiological function and undergo exquisite fine-tuning through multiple mechanisms. Here, we review known determinants and modulatory features of these two critical forms of channel regulation and their role in normal physiology and pathophysiology.
Collapse
Affiliation(s)
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Hussey JW, DeMarco E, DiSilvestre D, Brohus M, Busuioc AO, Iversen ED, Jensen HH, Nyegaard M, Overgaard MT, Ben-Johny M, Dick IE. Voltage Gated Calcium Channel Dysregulation May Contribute to Neurological Symptoms in Calmodulinopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626503. [PMID: 39677635 PMCID: PMC11642847 DOI: 10.1101/2024.12.02.626503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Calmodulinopathies are caused by mutations in calmodulin (CaM), and result in debilitating cardiac arrythmias such as long-QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT). In addition, many patients exhibit neurological comorbidities, including developmental delay and autism spectrum disorder. Until now, most work into these mutations has focused on cardiac effects, identifying impairment of Ca 2+ /CaM-dependent inactivation (CDI) of Ca V 1.2 channels as a major pathogenic mechanism. However, the impact of these mutations on neurological function has yet to be fully explored. CaM regulation of voltage-gated calcium channels (VGCCs) is a critical element of neuronal function, implicating multiple VGCC subtypes in the neurological pathogenesis of calmodulinopathies. Here, we explore the potential for pathological CaM variants to impair the Ca 2+ /CaM-dependent regulation of Ca V 1.3 and Ca V 2.1, both essential for neuronal function. We find that mutations in CaM can impair the CDI of Ca V 1.3 and reduce the Ca 2+ -dependent facilitation (CDF) of Ca V 2.1 channels. We find that mutations associated with significant neurological symptoms exhibit marked effects on Ca V 1.3 CDI, with overlapping but distinct impacts on Ca V 2.1 CDF. Moreover, while the majority of CaM variants demonstrated the ability to bind the IQ region of each channel, distinct differences were noted between Ca V 1.3 and Ca V 2.1, demonstrating distinct CaM interactions across the two channel subtypes. Further, C-domain CaM variants display a reduced ability to sense Ca 2+ when in complex with the Ca V IQ domains, explaining the Ca 2+ /CaM regulation deficits. Overall, these results support the possibility that disrupted Ca 2+ /CaM regulation of VGCCs may contribute to neurological pathogenesis of calmodulinopathies.
Collapse
|
4
|
Geng J, Yang Y, Li B, Yu Z, Qiu S, Zhang W, Gao S, Liu N, Liu Y, Wang B, Fan Y, Xing C, Liu X. Opto-chemogenetic inhibition of L-type Ca V1 channels in neurons through a membrane-assisted molecular linkage. CELL REPORTS METHODS 2024; 4:100898. [PMID: 39515337 PMCID: PMC11705922 DOI: 10.1016/j.crmeth.2024.100898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/28/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Genetically encoded inhibitors of CaV1 channels that operate via C-terminus-mediated inhibition (CMI) have been actively pursued. Here, we advance the design of CMI peptides by proposing a membrane-anchoring tag that is sufficient to link the inhibitory modules to the target channel as well as chemical and optogenetic modes of system control. We designed and implemented the constitutive and inducible CMI modules with appropriate dynamic ranges for the short and long variants of CaV1.3, both naturally occurring in neurons. Upon optical (near-infrared-responsive nanoparticles) and/or chemical (rapamycin) induction of FRB/FKBP binding, the designed peptides translocated onto the membrane via FRB-Ras, where the physical linkage requirement for CMI could be satisfied. The peptides robustly produced acute, potent, and specific inhibitions on both recombinant and neuronal CaV1 activities, including Ca2+ influx-neuritogenesis coupling. Validated through opto-chemogenetic induction, this prototype demonstrates Ca2+ channel modulation via membrane-assisted molecular linkage, promising broad applicability to diverse membrane proteins.
Collapse
Affiliation(s)
- Jinli Geng
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Yaxiong Yang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Boying Li
- School of Chemical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Zhen Yu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Shuang Qiu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Wen Zhang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Shixin Gao
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Nan Liu
- School of Life Sciences, Yunnan University, Kunming Yunnan 650091, China
| | - Yi Liu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Bo Wang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China.
| | - Chengfen Xing
- School of Chemical Engineering, Hebei University of Technology, Tianjin 300401, China.
| | - Xiaodong Liu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China.
| |
Collapse
|
5
|
Tang X, Ortner NJ, Nikonishyna YV, Fernández-Quintero ML, Kokot J, Striessnig J, Liedl KR. Pathogenicity of de novo CACNA1D Ca 2+ channel variants predicted from sequence co-variation. Eur J Hum Genet 2024; 32:1065-1073. [PMID: 38553610 PMCID: PMC11369236 DOI: 10.1038/s41431-024-01594-y] [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: 10/02/2023] [Revised: 03/02/2024] [Accepted: 03/12/2024] [Indexed: 09/04/2024] Open
Abstract
Voltage-gated L-type Cav1.3 Ca2+ channels support numerous physiological functions including neuronal excitability, sinoatrial node pacemaking, hearing, and hormone secretion. De novo missense mutations in the gene of their pore-forming α1-subunit (CACNA1D) induce severe gating defects which lead to autism spectrum disorder and a more severe neurological disorder with and without endocrine symptoms. The number of CACNA1D variants reported is constantly rising, but their pathogenic potential often remains unclear, which complicates clinical decision-making. Since functional tests are time-consuming and not always available, bioinformatic tools further improving pathogenicity potential prediction of novel variants are needed. Here we employed evolutionary analysis considering sequences of the Cav1.3 α1-subunit throughout the animal kingdom to predict the pathogenicity of human disease-associated CACNA1D missense variants. Co-variation analyses of evolutionary information revealed residue-residue couplings and allowed to generate a score, which correctly predicted previously identified pathogenic variants, supported pathogenicity in variants previously classified as likely pathogenic and even led to the re-classification or re-examination of 18 out of 80 variants previously assessed with clinical and electrophysiological data. Based on the prediction score, we electrophysiologically tested one variant (V584I) and found significant gating changes associated with pathogenic risks. Thus, our co-variation model represents a valuable addition to complement the assessment of the pathogenicity of CACNA1D variants completely independent of clinical diagnoses, electrophysiology, structural or biophysical considerations, and solely based on evolutionary analyses.
Collapse
Affiliation(s)
- Xuechen Tang
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Nadine J Ortner
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Yuliia V Nikonishyna
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Monica L Fernández-Quintero
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Janik Kokot
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria.
| | - Klaus R Liedl
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, A-6020, Innsbruck, Austria.
| |
Collapse
|
6
|
Thoreson WB, Zenisek D. Presynaptic Proteins and Their Roles in Visual Processing by the Retina. Annu Rev Vis Sci 2024; 10:347-375. [PMID: 38621251 PMCID: PMC11536687 DOI: 10.1146/annurev-vision-101322-111204] [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] [Indexed: 04/17/2024]
Abstract
The sense of vision begins in the retina, where light is detected and processed through a complex series of synaptic connections into meaningful information relayed to the brain via retinal ganglion cells. Light responses begin as tonic and graded signals in photoreceptors, later emerging from the retina as a series of spikes from ganglion cells. Processing by the retina extracts critical features of the visual world, including spatial frequency, temporal frequency, motion direction, color, contrast, and luminance. To achieve this, the retina has evolved specialized and unique synapse types. These include the ribbon synapses of photoreceptors and bipolar cells, the dendritic synapses of amacrine and horizontal cells, and unconventional synaptic feedback from horizontal cells to photoreceptors. We review these unique synapses in the retina with a focus on the presynaptic molecules and physiological properties that shape their capabilities.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Departments of Ophthalmology & Visual Sciences and Pharmacology & Experimental Neuroscience, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| | - David Zenisek
- Departments of Cellular and Molecular Physiology, Ophthalmology and Visual Sciences, and Neuroscience, Yale University, New Haven, Connecticut, USA;
| |
Collapse
|
7
|
Salamon S, Kuzmenkina E, Fried C, Matthes J. CaM-dependent modulation of human Ca V1.3 whole-cell and single-channel currents by C-terminal CaMKII phosphorylation site S1475. J Physiol 2024; 602:3955-3973. [PMID: 39037941 DOI: 10.1113/jp284972] [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: 05/12/2023] [Accepted: 07/02/2024] [Indexed: 07/24/2024] Open
Abstract
Phosphorylation enables rapid modulation of voltage-gated calcium channels (VGCC) in physiological and pathophysiological conditions. How phosphorylation modulates human CaV1.3 VGCC, however, is largely unexplored. We characterized modulation of CaV1.3 gating via S1475, the human equivalent of a phosphorylation site identified in the rat. S1475 is highly conserved in CaV1.3 but absent from all other high-voltage activating calcium channel types co-expressed with CaV1.3 in similar tissues. Further, it is located in the C-terminal EF-hand motif, which binds calmodulin (CaM). This is involved in calcium-dependent channel inactivation (CDI). We used amino acid exchanges that mimic either sustained phosphorylation (S1475D) or phosphorylation resistance (S1475A). Whole-cell and single-channel recordings of phosphorylation state imitating CaV1.3 variants in transiently transfected HEK-293 cells revealed functional relevance of S1475 in human CaV1.3. We obtained three main findings: (1) CaV1.3_S1475D, imitating sustained phosphorylation, displayed decreased current density, reduced CDI and (in-) activation kinetics shifted to more depolarized voltages compared with both wildtype CaV1.3 and the phosphorylation-resistant CaV1.3_S1475A variant. Corresponding to the decreased current density, we find a reduced open probability of CaV1.3_S1475D at the single-channel level. (2) Using CaM overexpression or depletion, we find that CaM is necessary for modulating CaV1.3 through S1475. (3) CaMKII activation led to CaV1.3_WT-current properties similar to those of CaV1.3_S1475D, but did not affect CaV1.3_S1475A, confirming that CaMKII modulates human CaV1.3 via S1475. Given the physiological and pathophysiological importance of CaV1.3, our findings on the S1475-mediated interplay of phosphorylation, CaM interaction and CDI provide hints for approaches on specific CaV1.3 modulation under physiological and pathophysiological conditions. KEY POINTS: Phosphorylation modulates activity of voltage-gated L-type calcium channels for specific cellular needs but is largely unexplored for human CaV1.3 channels. Here we report that S1475, a CaMKII phosphorylation site identified in rats, is functionally relevant in human CaV1.3. Imitating phosphorylation states at S1475 alters current density and inactivation in a calmodulin-dependent manner. In wildtype CaV1.3 but not in the phosphorylation-resistant variant S1475A, CaMKII activation elicits effects similar to constitutively mimicking phosphorylation at S1475. Our findings provide novel insights on the interplay of modulatory mechanisms of human CaV1.3 channels, and present a possible target for CaV1.3-specific gating modulation in physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Sarah Salamon
- Center of Pharmacology, Institute II, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Elza Kuzmenkina
- Center of Pharmacology, Institute II, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Cora Fried
- Center of Pharmacology, Institute II, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jan Matthes
- Center of Pharmacology, Institute II, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
8
|
Lebedeva M, Kubištová A, Spišská V, Filipovská E, Pačesová D, Svobodová I, Kuchtiak V, Balík A, Bendová Z. The disruption of circadian rhythmicity of gene expression in the hippocampus and associated structures in Gria2 R/R mice; a comparison with C57BL/6J and Adar2 -/- mice strains. Brain Res 2024; 1826:148739. [PMID: 38157956 DOI: 10.1016/j.brainres.2023.148739] [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: 08/21/2023] [Revised: 11/18/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Adar2-/- mice are a widely used model for studying the physiological consequences of reduced RNA editing. These mice are viable only when the Q/R editing site of the Gria2 subunit of the AMPA receptor is constitutively mutated to the codon for arginine, and Gria2R/R mice often serve as the sole control for Adar2-/- mice. Our study aimed to investigate whether ADAR2 inactivity and the Gria2R/R phenotype affect the rhythmicity of the circadian clock gene pattern and the expression of Gria1 and Gria2 subunits in the suprachiasmatic nucleus (SCN), hippocampus, parietal cortex and liver. Our data show that Gria2R/R mice completely lost circadian rhythmicity in the hippocampus compared to Adar2-/- mice. Compared to C57BL/6J mice, the expression profiles in the hippocampus and parietal cortex of Gria2R/R mice differ to the same extent as in Adar2-/-. No alterations were detected in the circadian profiles in the livers. These data suggest that the natural gradual postnatal increase in the editing of the Q/R site of the Gria2 subunit may be important for the development of circadian clockwork in some brain structures, and the use of Gria2R/R mice as the only control to Adar2-/- mice in the experiments dependent on the hippocampus and parietal cortex should therefore be considered.
Collapse
Affiliation(s)
- Maria Lebedeva
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic; National Institute of Mental Health, Klecany, Czech Republic
| | - Aneta Kubištová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Veronika Spišská
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Eva Filipovská
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Dominika Pačesová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Irena Svobodová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Viktor Kuchtiak
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Balík
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | - Zdeňka Bendová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic; National Institute of Mental Health, Klecany, Czech Republic.
| |
Collapse
|
9
|
Yang Q, Perfitt TL, Quay J, Hu L, Lawson-Qureshi D, Colbran RJ. Clustering of Ca V 1.3 L-type calcium channels by Shank3. J Neurochem 2023; 167:16-37. [PMID: 37392026 PMCID: PMC10543641 DOI: 10.1111/jnc.15880] [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: 10/14/2022] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 07/02/2023]
Abstract
Clustering of L-type voltage-gated Ca2+ channels (LTCCs) in the plasma membrane is increasingly implicated in creating highly localized Ca2+ signaling nanodomains. For example, neuronal LTCC activation can increase phosphorylation of the nuclear CREB transcription factor by increasing Ca2+ concentrations within a nanodomain close to the channel, without requiring bulk Ca2+ increases in the cytosol or nucleus. However, the molecular basis for LTCC clustering is poorly understood. The postsynaptic scaffolding protein Shank3 specifically associates with one of the major neuronal LTCCs, the CaV 1.3 calcium channel, and is required for optimal LTCC-dependent excitation-transcription coupling. Here, we co-expressed CaV 1.3 α1 subunits with two distinct epitope-tags with or without Shank3 in HEK cells. Co-immunoprecipitation studies using the cell lysates revealed that Shank3 can assemble complexes containing multiple CaV 1.3 α1 subunits under basal conditions. Moreover, CaV 1.3 LTCC complex formation was facilitated by CaV β subunits (β3 and β2a), which also interact with Shank3. Shank3 interactions with CaV 1.3 LTCCs and multimeric CaV 1.3 LTCC complex assembly were disrupted following the addition of Ca2+ to cell lysates, perhaps simulating conditions within an activated CaV 1.3 LTCC nanodomain. In intact HEK293T cells, co-expression of Shank3 enhanced the intensity of membrane-localized CaV 1.3 LTCC clusters under basal conditions, but not after Ca2+ channel activation. Live cell imaging studies also revealed that Ca2+ influx through LTCCs disassociated Shank3 from CaV 1.3 LTCCs clusters and reduced the CaV 1.3 cluster intensity. Deletion of the Shank3 PDZ domain prevented both binding to CaV 1.3 and the changes in multimeric CaV 1.3 LTCC complex assembly in vitro and in HEK293 cells. Finally, we found that shRNA knock-down of Shank3 expression in cultured rat primary hippocampal neurons reduced the intensity of surface-localized CaV 1.3 LTCC clusters in dendrites. Taken together, our findings reveal a novel molecular mechanism contributing to neuronal LTCC clustering under basal conditions.
Collapse
Affiliation(s)
- Qian Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Tyler L. Perfitt
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
- Current address: Rare Disease Research Unit, Pfizer Inc
| | - Juliana Quay
- Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Lan Hu
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Dorian Lawson-Qureshi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Roger J. Colbran
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
- Vanderbilt-Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| |
Collapse
|
10
|
Török F, Tezcan K, Filippini L, Fernández-Quintero ML, Zanetti L, Liedl KR, Drexel RS, Striessnig J, Ortner NJ. Germline de novo variant F747S extends the phenotypic spectrum of CACNA1D Ca2+ channelopathies. Hum Mol Genet 2023; 32:847-859. [PMID: 36208199 PMCID: PMC9941835 DOI: 10.1093/hmg/ddac248] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/22/2022] [Accepted: 09/30/2022] [Indexed: 11/14/2022] Open
Abstract
Germline gain-of-function missense variants in the pore-forming Cav1.3 α1-subunit (CACNA1D gene) confer high risk for a severe neurodevelopmental disorder with or without endocrine symptoms. Here, we report a 4-week-old new-born with the novel de novo missense variant F747S with a so far not described prominent jittering phenotype in addition to symptoms previously reported for CACNA1D mutations including developmental delay, elevated aldosterone level and transient hypoglycemia. We confirmed the pathogenicity of this variant in whole-cell patch-clamp experiments with wild-type and F747S mutant channels heterologously expressed together with α2δ1 and cytosolic β3 or membrane-bound β2a subunits. Mutation F747S caused the quantitatively largest shift in the voltage dependence of activation (-28 mV) reported so far for CACNA1D germline mutations. It also shifted inactivation to more negative voltages, slowed the time course of current inactivation and slowed current deactivation upon repolarization with both co-expressed β-subunits. In silico modelling and molecular docking, simulations revealed that this gain-of-function phenotype can be explained by formation of a novel inter-domain hydrogen bond between mutant residues S747 (IIS6) with N1145 (IIIS6) stabilizing selectively the activated open channel state. F747S displayed 2-6-fold increased sensitivity for the L-type Ca2+ channel blocker isradipine compared to wild type. Our data confirm the pathogenicity of the F747S variant with very strong gain-of-function gating changes, which may contribute to the novel jittering phenotype. Increased sensitivity for isradipine suggests this drug for potential symptomatic off-label treatment for carriers of this mutation.
Collapse
Affiliation(s)
- Ferenc Török
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Kamer Tezcan
- Department of Genetics, Kaiser Permanente, Sacramento, CA 95825, USA
| | - Ludovica Filippini
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Monica L Fernández-Quintero
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Lucia Zanetti
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Klaus R Liedl
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Raphaela S Drexel
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Nadine J Ortner
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
11
|
Abstract
Tightly controlled Ca2+ influx through voltage-gated Ca2+ channels (Cavs) is indispensable for proper physiological function. Thus, it is not surprising that Cav loss and/or gain of function have been implicated in human pathology. Deficiency of Cav1.3 L-type Ca2+ channels (LTCCs) causes deafness and bradycardia, whereas several genetic variants of CACNA1D, the gene encoding the pore-forming α1 subunit of Cav1.3, have been linked to various disease phenotypes, such as hypertension, congenital hypoglycemia, or autism. These variants include not only common polymorphisms associated with an increased disease risk, but also rare de novo missense variants conferring high risk. This review provides a concise summary of disease-associated CACNA1D variants, whereas the main focus lies on de novo germline variants found in individuals with a neurodevelopmental disorder of variable severity. Electrophysiological recordings revealed activity-enhancing gating changes induced by these de novo variants, and tools to predict their pathogenicity and to study the resulting pathophysiological consequences will be discussed. Despite the low number of affected patients, potential phenotype-genotype correlations and factors that could impact the severity of symptoms will be covered. Since increased channel activity is assumed as the disease-underlying mechanism, pharmacological inhibition could be a treatment option. In the absence of Cav1.3-selective blockers, dihydropyridine LTCC inhibitors clinically approved for the treatment of hypertension may be used for personalized off-label trials. Findings from in vitro studies and treatment attempts in some of the patients seem promising as outlined. Taken together, due to advances in diagnostic sequencing techniques the number of reported CACNA1D variants in human diseases is constantly rising. Evidence from in silico, in vitro, and in vivo disease models can help to predict the pathogenic potential of such variants and to guide diagnosis and treatment in the clinical practice when confronted with patients harboring CACNA1D variants.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
12
|
Zaveri S, Srivastava U, Qu YS, Chahine M, Boutjdir M. Pathophysiology of Ca v1.3 L-type calcium channels in the heart. Front Physiol 2023; 14:1144069. [PMID: 37025382 PMCID: PMC10070707 DOI: 10.3389/fphys.2023.1144069] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Ca2+ plays a crucial role in excitation-contraction coupling in cardiac myocytes. Dysfunctional Ca2+ regulation alters the force of contraction and causes cardiac arrhythmias. Ca2+ entry into cardiomyocytes is mediated mainly through L-type Ca2+ channels, leading to the subsequent Ca2+ release from the sarcoplasmic reticulum. L-type Ca2+ channels are composed of the conventional Cav1.2, ubiquitously expressed in all heart chambers, and the developmentally regulated Cav1.3, exclusively expressed in the atria, sinoatrial node, and atrioventricular node in the adult heart. As such, Cav1.3 is implicated in the pathogenesis of sinoatrial and atrioventricular node dysfunction as well as atrial fibrillation. More recently, Cav1.3 de novo expression was suggested in heart failure. Here, we review the functional role, expression levels, and regulation of Cav1.3 in the heart, including in the context of cardiac diseases. We believe that the elucidation of the functional and molecular pathways regulating Cav1.3 in the heart will assist in developing novel targeted therapeutic interventions for the aforementioned arrhythmias.
Collapse
Affiliation(s)
- Sahil Zaveri
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, NY, United States
| | - Ujala Srivastava
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
| | - Yongxia Sarah Qu
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, NY, United States
- Department of Cardiology, New York Presbyterian Brooklyn Methodist Hospital, New York, NY, United States
| | - Mohamed Chahine
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Québec, QC, Canada
- Department of Medicine, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, NY, United States
- Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, United States
- *Correspondence: Mohamed Boutjdir,
| |
Collapse
|
13
|
Rinné S, Stallmeyer B, Pinggera A, Netter MF, Matschke LA, Dittmann S, Kirchhefer U, Neudorf U, Opp J, Striessnig J, Decher N, Schulze-Bahr E. Whole Exome Sequencing Identifies a Heterozygous Variant in the Cav1.3 Gene CACNA1D Associated with Familial Sinus Node Dysfunction and Focal Idiopathic Epilepsy. Int J Mol Sci 2022; 23:14215. [PMID: 36430690 PMCID: PMC9693521 DOI: 10.3390/ijms232214215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Cav1.3 voltage-gated L-type calcium channels (LTCCs) are involved in cardiac pacemaking, hearing and hormone secretion, but are also expressed postsynaptically in neurons. So far, homozygous loss of function mutations in CACNA1D encoding the Cav1.3 α1-subunit are described in congenital sinus node dysfunction and deafness. In addition, germline mutations in CACNA1D have been linked to neurodevelopmental syndromes including epileptic seizures, autism, intellectual disability and primary hyperaldosteronism. Here, a three-generation family with a syndromal phenotype of sinus node dysfunction, idiopathic epilepsy and attention deficit hyperactivity disorder (ADHD) is investigated. Whole genome sequencing and functional heterologous expression studies were used to identify the disease-causing mechanisms in this novel syndromal disorder. We identified a heterozygous non-synonymous variant (p.Arg930His) in the CACNA1D gene that cosegregated with the combined clinical phenotype in an autosomal dominant manner. Functional heterologous expression studies showed that the CACNA1D variant induces isoform-specific alterations of Cav1.3 channel gating: a gain of ion channel function was observed in the brain-specific short CACNA1D isoform (Cav1.3S), whereas a loss of ion channel function was seen in the long (Cav1.3L) isoform. The combined gain-of-function (GOF) and loss-of-function (LOF) induced by the R930H variant are likely to be associated with the rare combined clinical and syndromal phenotypes in the family. The GOF in the Cav1.3S variant with high neuronal expression is likely to result in epilepsy, whereas the LOF in the long Cav1.3L variant results in sinus node dysfunction.
Collapse
Affiliation(s)
- Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Birgit Stallmeyer
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Muenster, 48149 Muenster, Germany
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Michael F. Netter
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Lina A. Matschke
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Sven Dittmann
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Muenster, 48149 Muenster, Germany
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University Hospital Muenster, 48149 Muenster, Germany
| | - Ulrich Neudorf
- Zentrum für Kinder-und Jugendmedizin, Klinik für Kinderheilkunde III-Bereich Kardiologie, University Hospital Essen, 45147 Essen, Germany
| | - Joachim Opp
- Ev. Krankenhaus Oberhausen, 46047 Oberhausen, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Muenster, 48149 Muenster, Germany
| |
Collapse
|
14
|
Metzner C, Mäki-Marttunen T, Karni G, McMahon-Cole H, Steuber V. The effect of alterations of schizophrenia-associated genes on gamma band oscillations. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:46. [PMID: 35854005 PMCID: PMC9261091 DOI: 10.1038/s41537-022-00255-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
Abnormalities in the synchronized oscillatory activity of neurons in general and, specifically in the gamma band, might play a crucial role in the pathophysiology of schizophrenia. While these changes in oscillatory activity have traditionally been linked to alterations at the synaptic level, we demonstrate here, using computational modeling, that common genetic variants of ion channels can contribute strongly to this effect. Our model of primary auditory cortex highlights multiple schizophrenia-associated genetic variants that reduce gamma power in an auditory steady-state response task. Furthermore, we show that combinations of several of these schizophrenia-associated variants can produce similar effects as the more traditionally considered synaptic changes. Overall, our study provides a mechanistic link between schizophrenia-associated common genetic variants, as identified by genome-wide association studies, and one of the most robust neurophysiological endophenotypes of schizophrenia.
Collapse
Affiliation(s)
- Christoph Metzner
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany.
- Biocomputation Research Group, School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom.
| | | | - Gili Karni
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany
- Minerva Schools at KGI, San Francisco, CA, USA
| | - Hana McMahon-Cole
- Neural Information Processing Group, Institute of Software Engineering and Theoretical Computer Science, Technische Universität Berlin, Berlin, Germany
- Minerva Schools at KGI, San Francisco, CA, USA
| | - Volker Steuber
- Biocomputation Research Group, School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
15
|
Hofer NT, Pinggera A, Nikonishyna YV, Tuluc P, Fritz EM, Obermair GJ, Striessnig J. Stabilization of negative activation voltages of Cav1.3 L-Type Ca 2+-channels by alternative splicing. Channels (Austin) 2021; 15:38-52. [PMID: 33380256 PMCID: PMC7781618 DOI: 10.1080/19336950.2020.1859260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 11/30/2022] Open
Abstract
-->Low voltage-activated Cav1.3 L-type Ca2+-channels are key regulators of neuronal excitability controlling neuronal development and different types of learning and memory. Their physiological functions are enabled by their negative activation voltage-range, which allows Cav1.3 to be active at subthreshold voltages. Alternative splicing in the C-terminus of their pore-forming α1-subunits gives rise to C-terminal long (Cav1.3L) and short (Cav1.3S) splice variants allowing Cav1.3S to activate at even more negative voltages than Cav1.3L. We discovered that inclusion of exons 8b, 11, and 32 in Cav1.3S further shifts activation (-3 to -4 mV) and inactivation (-4 to -6 mV) to more negative voltages as revealed by functional characterization in tsA-201 cells. We found transcripts of these exons in mouse chromaffin cells, the cochlea, and the brain. Our data further suggest that Cav1.3-containing exons 11 and 32 constitute a significant part of native channels in the brain. We therefore investigated the effect of these splice variants on human disease variants. Splicing did not prevent the gating defects of the previously reported human pathogenic variant S652L, which further shifted the voltage-dependence of activation of exon 11-containing channels by more than -12 mV. In contrast, we found no evidence for gating changes of the CACNA1D missense variant R498L, located in exon 11, which has recently been identified in a patient with an epileptic syndrome. Our data demonstrate that alternative splicing outside the C-terminus involving exons 11 and 32 contributes to channel fine-tuning by stabilizing negative activation and inactivation gating properties of wild-type and mutant Cav1.3 channels.
Collapse
Affiliation(s)
- Nadja T. Hofer
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Yuliia V. Nikonishyna
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Eva M. Fritz
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Gerald J. Obermair
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| |
Collapse
|
16
|
Jiang MC, Birch DV, Heckman CJ, Tysseling VM. The Involvement of Ca V1.3 Channels in Prolonged Root Reflexes and Its Potential as a Therapeutic Target in Spinal Cord Injury. Front Neural Circuits 2021; 15:642111. [PMID: 33867945 PMCID: PMC8044857 DOI: 10.3389/fncir.2021.642111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) results in not only the loss of voluntary muscle control, but also in the presence of involuntary movement or spasms. These spasms post-SCI involve hyperexcitability in the spinal motor system. Hyperactive motor commands post SCI result from enhanced excitatory postsynaptic potentials (EPSPs) and persistent inward currents in voltage-gated L-type calcium channels (LTCCs), which are reflected in evoked root reflexes with different timings. To further understand the contributions of these cellular mechanisms and to explore the involvement of LTCC subtypes in SCI-induced hyperexcitability, we measured root reflexes with ventral root recordings and motoneuron activities with intracellular recordings in an in vitro preparation using a mouse model of chronic SCI (cSCI). Specifically, we explored the effects of 1-(3-chlorophenethyl)-3-cyclopentylpyrimidine-2,4,6-(1H,3H,5H)-trione (CPT), a selective negative allosteric modulator of CaV1.3 LTCCs. Our results suggest a hyperexcitability in the spinal motor system in these SCI mice. Bath application of CPT displayed slow onset but dose-dependent inhibition of the root reflexes with the strongest effect on LLRs. However, the inhibitory effect of CPT is less potent in cSCI mice than in acute SCI (aSCI) mice, suggesting changes either in composition of CaV1.3 or other cellular mechanisms in cSCI mice. For intracellular recordings, the intrinsic plateau potentials, was observed in more motoneurons in cSCI mice than in aSCI mice. CPT inhibited the plateau potentials and reduced motoneuron firings evoked by intracellular current injection. These results suggest that the LLR is an important target and that CPT has potential in the therapy of SCI-induced muscle spasms.
Collapse
Affiliation(s)
- Mingchen C Jiang
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Derin V Birch
- Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Charles J Heckman
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Vicki M Tysseling
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
17
|
Ortner NJ. Voltage-Gated Ca 2+ Channels in Dopaminergic Substantia Nigra Neurons: Therapeutic Targets for Neuroprotection in Parkinson's Disease? Front Synaptic Neurosci 2021; 13:636103. [PMID: 33716705 PMCID: PMC7952618 DOI: 10.3389/fnsyn.2021.636103] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
The loss of dopamine (DA)-producing neurons in the substantia nigra pars compacta (SN) underlies the core motor symptoms of the progressive movement disorder Parkinson's disease (PD). To date, no treatment to prevent or slow SN DA neurodegeneration exists; thus, the identification of the underlying factors contributing to the high vulnerability of these neurons represents the basis for the development of novel therapies. Disrupted Ca2+ homeostasis and mitochondrial dysfunction seem to be key players in the pathophysiology of PD. The autonomous pacemaker activity of SN DA neurons, in combination with low cytosolic Ca2+ buffering, leads to large somatodendritic fluctuations of intracellular Ca2+ levels that are linked to elevated mitochondrial oxidant stress. L-type voltage-gated Ca2+ channels (LTCCs) contribute to these Ca2+ oscillations in dendrites, and LTCC inhibition was beneficial in cellular and in vivo animal models of PD. However, in a recently completed phase 3 clinical trial, the dihydropyridine (DHP) LTCC inhibitor isradipine failed to slow disease progression in early PD patients, questioning the feasibility of DHPs for PD therapy. Novel evidence also suggests that R- and T-type Ca2+ channels (RTCCs and TTCCs, respectively) represent potential PD drug targets. This short review aims to (re)evaluate the therapeutic potential of LTCC, RTCC, and TTCC inhibition in light of novel preclinical and clinical data and the feasibility of available Ca2+ channel blockers to modify PD disease progression. I also summarize their cell-specific roles for SN DA neuron function and describe how their gating properties allow activity (and thus their contribution to stressful Ca2+ oscillations) during pacemaking.
Collapse
Affiliation(s)
- Nadine J. Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
18
|
Taylor NJ, Gaynanova I, Eschrich SA, Welsh EA, Garrett TJ, Beecher C, Sharma R, Koomen JM, Smalley KSM, Messina JL, Kanetsky PA. Metabolomics of primary cutaneous melanoma and matched adjacent extratumoral microenvironment. PLoS One 2020; 15:e0240849. [PMID: 33108391 PMCID: PMC7591037 DOI: 10.1371/journal.pone.0240849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Melanoma causes the vast majority of deaths attributable to skin cancer, largely due to its propensity for metastasis. To date, few studies have examined molecular changes between primary cutaneous melanoma and adjacent putatively normal skin. To broaden temporal inferences related to initiation of disease, we performed a metabolomics investigation of primary melanoma and matched extratumoral microenvironment (EM) tissues; and, to make inferences about progressive disease, we also compared unmatched metastatic melanoma tissues to EM tissues. METHODS Ultra-high performance liquid chromatography-mass spectrometry-based metabolic profiling was performed on frozen human tissues. RESULTS We observed 824 metabolites as differentially abundant among 33 matched tissue samples, and 1,118 metabolites as differentially abundant between metastatic melanoma (n = 46) and EM (n = 34) after false discovery rate (FDR) adjustment (p<0.01). No significant differences in metabolite abundances were noted comparing primary and metastatic melanoma tissues. CONCLUSIONS Overall, pathway-based results significantly distinguished melanoma tissues from EM in the metabolism of: ascorbate and aldarate, propanoate, tryptophan, histidine, and pyrimidine. Within pathways, the majority of individual metabolite abundances observed in comparisons of primary melanoma vs. EM and metastatic melanoma vs. EM were directionally consistent. This observed concordance suggests most identified compounds are implicated in the initiation or maintenance of melanoma.
Collapse
Affiliation(s)
- Nicholas J. Taylor
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas, United States of America
| | - Irina Gaynanova
- Department of Statistics, Texas A&M University, College Station, Texas, United States of America
| | - Steven A. Eschrich
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Eric A. Welsh
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Timothy J. Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Chris Beecher
- IROA Technologies, Chapel Hill, North Carolina, United States of America
| | - Ritin Sharma
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - John M. Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Keiran S. M. Smalley
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Jane L. Messina
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Peter A. Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| |
Collapse
|
19
|
Verma A, Ravindranath V. Ca V1.3 L-Type Calcium Channels Increase the Vulnerability of Substantia Nigra Dopaminergic Neurons in MPTP Mouse Model of Parkinson's Disease. Front Aging Neurosci 2020; 11:382. [PMID: 32009942 PMCID: PMC6978652 DOI: 10.3389/fnagi.2019.00382] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022] Open
Abstract
Mechanisms underlying the selective vulnerability of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) over those in the ventral tegmental area (VTA) to degeneration in Parkinson’s disease (PD) remain poorly understood. DA neurons of SNpc and VTA are autonomous pacemakers but pacemaking in SNpc but not in VTA is accompanied by calcium influx through L-type calcium channel, CaV1.3 contributing to increased intracellular calcium and hence to cell death. CaV1.342A, an alternatively spliced short variant of CaV1.3 has increased calcium influx. We, therefore studied the role of CaV1.342 (full-length channel) and CaV1.342A in mouse SNpc in PD pathogenesis by quantifying mRNA levels of CaV1.342 and CaV1.342A in SNpc and followed the change in their levels in MPTP induced parkinsonism mouse model. Using in situ hybridization and immunohistochemistry we observed the localization of mRNA of CaV1.342 and CaV1.342A in tyrosine hydroxylase (TH) positive DA neurons. Further, mRNA levels of CaV1.342A were higher in SNpc as compared to the cortex. Upon MPTP treatment, mRNA levels of CaV1.342 and CaV1.342A maintained their levels in SNpc in spite of the loss of ~50% of the DA neurons. This indicates that the expression of CaV1.342 and CaV1.342A is maintained at a robust level during the degenerative process in the parkinsonism model.
Collapse
Affiliation(s)
- Aditi Verma
- Centre for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Bangalore, India.,Centre for Brain Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
20
|
Ortner NJ, Pinggera A, Hofer NT, Siller A, Brandt N, Raffeiner A, Vilusic K, Lang I, Blum K, Obermair GJ, Stefan E, Engel J, Striessnig J. RBP2 stabilizes slow Cav1.3 Ca 2+ channel inactivation properties of cochlear inner hair cells. Pflugers Arch 2019; 472:3-25. [PMID: 31848688 PMCID: PMC6960213 DOI: 10.1007/s00424-019-02338-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 12/04/2019] [Indexed: 01/31/2023]
Abstract
Cav1.3 L-type Ca2+ channels (LTCCs) in cochlear inner hair cells (IHCs) are essential for hearing as they convert sound-induced graded receptor potentials into tonic postsynaptic glutamate release. To enable fast and indefatigable presynaptic Ca2+ signaling, IHC Cav1.3 channels exhibit a negative activation voltage range and uniquely slow inactivation kinetics. Interaction with CaM-like Ca2+-binding proteins inhibits Ca2+-dependent inactivation, while the mechanisms underlying slow voltage-dependent inactivation (VDI) are not completely understood. Here we studied if the complex formation of Cav1.3 LTCCs with the presynaptic active zone proteins RIM2α and RIM-binding protein 2 (RBP2) can stabilize slow VDI. We detected both RIM2α and RBP isoforms in adult mouse IHCs, where they co-localized with Cav1.3 and synaptic ribbons. Using whole-cell patch-clamp recordings (tsA-201 cells), we assessed their effect on the VDI of the C-terminal full-length Cav1.3 (Cav1.3L) and a short splice variant (Cav1.342A) that lacks the C-terminal RBP2 interaction site. When co-expressed with the auxiliary β3 subunit, RIM2α alone (Cav1.342A) or RIM2α/RBP2 (Cav1.3L) reduced Cav1.3 VDI to a similar extent as observed in IHCs. Membrane-anchored β2 variants (β2a, β2e) that inhibit inactivation on their own allowed no further modulation of inactivation kinetics by RIM2α/RBP2. Moreover, association with RIM2α and/or RBP2 consolidated the negative Cav1.3 voltage operating range by shifting the channel's activation threshold toward more hyperpolarized potentials. Taken together, the association with "slow" β subunits (β2a, β2e) or presynaptic scaffolding proteins such as RIM2α and RBP2 stabilizes physiological gating properties of IHC Cav1.3 LTCCs in a splice variant-dependent manner ensuring proper IHC function.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Nadja T Hofer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Anita Siller
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Niels Brandt
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Andrea Raffeiner
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Kristina Vilusic
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Isabelle Lang
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Gerald J Obermair
- Division of Physiology, Medical University Innsbruck, Innsbruck, Austria.,Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Eduard Stefan
- Institute of Biochemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Jutta Engel
- Department of Biophysics and CIPMM, Saarland University, Homburg, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
21
|
Mäki-Marttunen T, Devor A, Phillips WA, Dale AM, Andreassen OA, Einevoll GT. Computational Modeling of Genetic Contributions to Excitability and Neural Coding in Layer V Pyramidal Cells: Applications to Schizophrenia Pathology. Front Comput Neurosci 2019; 13:66. [PMID: 31616272 PMCID: PMC6775251 DOI: 10.3389/fncom.2019.00066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 09/09/2019] [Indexed: 11/13/2022] Open
Abstract
Pyramidal cells in layer V of the neocortex are one of the most widely studied neuron types in the mammalian brain. Due to their role as integrators of feedforward and cortical feedback inputs, they are well-positioned to contribute to the symptoms and pathology in mental disorders-such as schizophrenia-that are characterized by a mismatch between the internal perception and external inputs. In this modeling study, we analyze the input/output properties of layer V pyramidal cells and their sensitivity to modeled genetic variants in schizophrenia-associated genes. We show that the excitability of layer V pyramidal cells and the way they integrate inputs in space and time are altered by many types of variants in ion-channel and Ca2+ transporter-encoding genes that have been identified as risk genes by recent genome-wide association studies. We also show that the variability in the output patterns of spiking and Ca2+ transients in layer V pyramidal cells is altered by these model variants. Importantly, we show that many of the predicted effects are robust to noise and qualitatively similar across different computational models of layer V pyramidal cells. Our modeling framework reveals several aspects of single-neuron excitability that can be linked to known schizophrenia-related phenotypes and existing hypotheses on disease mechanisms. In particular, our models predict that single-cell steady-state firing rate is positively correlated with the coding capacity of the neuron and negatively correlated with the amplitude of a prepulse-mediated adaptation and sensitivity to coincidence of stimuli in the apical dendrite and the perisomatic region of a layer V pyramidal cell. These results help to uncover the voltage-gated ion-channel and Ca2+ transporter-associated genetic underpinnings of schizophrenia phenotypes and biomarkers.
Collapse
Affiliation(s)
| | - Anna Devor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.,Department of Radiology, University of California San Diego, La Jolla, CA, United States.,Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - William A Phillips
- Psychology, Faculty of Natural Sciences, University of Stirling, Stirling, United Kingdom
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.,Department of Radiology, University of California San Diego, La Jolla, CA, United States
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gaute T Einevoll
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Pinggera A, Negro G, Tuluc P, Brown MJ, Lieb A, Striessnig J. Gating defects of disease-causing de novo mutations in Ca v1.3 Ca 2+ channels. Channels (Austin) 2019; 12:388-402. [PMID: 30465465 PMCID: PMC6287693 DOI: 10.1080/19336950.2018.1546518] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Recently, we and others identified somatic and germline de novo gain-of-function mutations in CACNA1D, the gene encoding the α1-subunit of voltage-gated Cav1.3 Ca2+-channels. While somatic mutations identified in aldosterone producing adenomas (APAs) underlie treatment-resistant hypertension, germline CACNA1D mutations are associated with a neurodevelopmental disorder characterized by a wide symptomatic spectrum, including autism spectrum disorder. The number of newly identified CACNA1D missense mutations is constantly growing, but their pathogenic potential is difficult to predict in silico, making functional studies indispensable to assess their contribution to disease risk. Here we report the functional characterization of previously identified CACNA1D APA mutations F747L and M1354I using whole-cell patch-clamp electrophysiology upon recombinant expression in tsA-201 cells. We also investigated if alternative splicing of Cav1.3 affects the aberrant gating of the previously characterized APA mutation R990H and two mutations associated with autism spectrum disorder (A479G and G407R). Splice-variant dependent gating changes are of particular interest for germline mutations, since the relative expression of Cav1.3 splice variants differs across different tissues and within brain regions and might therefore result in tissue-specific phenotypes. Our data revealed a complex gain-of-function phenotype for APA mutation F747L confirming its pathogenic role. Furthermore, we found splice-variant dependent gating changes in R990H, A749G and G407R. M1354I did not change channel function of Cav1.3 splice variants and should therefore be considered a rare non-pathogenic variant until further proof for its pathogenicity is obtained. Our new findings together with previously published data allow classification of pathogenic CACNA1D mutations into four categories based on prototypical functional changes.
Collapse
Affiliation(s)
- Alexandra Pinggera
- a Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences , University of Innsbruck , Innsbruck , Austria
| | - Giulia Negro
- a Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences , University of Innsbruck , Innsbruck , Austria
| | - Petronel Tuluc
- a Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences , University of Innsbruck , Innsbruck , Austria
| | - Morris J Brown
- b William Harvey Research Institute , Queen Mary University of London , London , UK
| | - Andreas Lieb
- a Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences , University of Innsbruck , Innsbruck , Austria
| | - Jörg Striessnig
- a Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences , University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
23
|
Intrinsic planar polarity mechanisms influence the position-dependent regulation of synapse properties in inner hair cells. Proc Natl Acad Sci U S A 2019; 116:9084-9093. [PMID: 30975754 DOI: 10.1073/pnas.1818358116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Encoding the wide range of audible sounds in the mammalian cochlea is collectively achieved by functionally diverse type I spiral ganglion neurons (SGNs) at each tonotopic position. The firing of each SGN is thought to be driven by an individual active zone (AZ) of a given inner hair cell (IHC). These AZs present distinct properties according to their position within the IHC, to some extent forming a gradient between the modiolar and the pillar IHC side. In this study, we investigated whether signaling involved in planar polarity at the apical surface can influence position-dependent AZ properties at the IHC base. Specifically, we tested the role of Gαi proteins and their binding partner LGN/Gpsm2 implicated in cytoskeleton polarization and hair cell (HC) orientation along the epithelial plane. Using high and superresolution immunofluorescence microscopy as well as patch-clamp combined with confocal Ca2+ imaging we analyzed IHCs in which Gαi signaling was blocked by Cre-induced expression of the pertussis toxin catalytic subunit (PTXa). PTXa-expressing IHCs exhibited larger CaV1.3 Ca2+-channel clusters and consequently greater Ca2+ influx at the whole-cell and single-synapse levels, which also showed a hyperpolarized shift of activation. Moreover, PTXa expression collapsed the modiolar-pillar gradients of ribbon size and maximal synaptic Ca2+ influx. Finally, genetic deletion of Gαi3 and LGN/Gpsm2 also disrupted the modiolar-pillar gradient of ribbon size. We propose a role for Gαi proteins and LGN in regulating the position-dependent AZ properties in IHCs and suggest that this signaling pathway contributes to setting up the diverse firing properties of SGNs.
Collapse
|
24
|
Single-Channel Resolution of the Interaction between C-Terminal Ca V1.3 Isoforms and Calmodulin. Biophys J 2019; 116:836-846. [PMID: 30773296 DOI: 10.1016/j.bpj.2019.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/05/2019] [Accepted: 01/16/2019] [Indexed: 12/21/2022] Open
Abstract
Voltage-dependent calcium (CaV) 1.3 channels are involved in the control of cellular excitability and pacemaking in neuronal, cardiac, and sensory cells. Various proteins interact with the alternatively spliced channel C-terminus regulating gating of CaV1.3 channels. Binding of a regulatory calcium-binding protein calmodulin (CaM) to the proximal C-terminus leads to the boosting of channel activity and promotes calcium-dependent inactivation (CDI). The C-terminal modulator domain (CTM) of CaV1.3 channels can interfere with the CaM binding, thereby inhibiting channel activity and CDI. Here, we compared single-channel gating behavior of two natural CaV1.3 splice isoforms: the long CaV1.342 with the full-length CTM and the short CaV1.342A with the C-terminus truncated before the CTM. We found that CaM regulation of CaV1.3 channels is dynamic on a minute timescale. We observed that at equilibrium, single CaV1.342 channels occasionally switched from low to high open probability, which perhaps reflects occasional binding of CaM despite the presence of CTM. Similarly, when the amount of the available CaM in the cell was reduced, the short CaV1.342A isoform showed patterns of the low channel activity. CDI also underwent periodic changes with corresponding kinetics in both isoforms. Our results suggest that the competition between CTM and CaM is influenced by calcium, allowing further fine-tuning of CaV1.3 channel activity for particular cellular needs.
Collapse
|
25
|
A Selectivity Filter Gate Controls Voltage-Gated Calcium Channel Calcium-Dependent Inactivation. Neuron 2019; 101:1134-1149.e3. [DOI: 10.1016/j.neuron.2019.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/12/2018] [Accepted: 12/31/2018] [Indexed: 11/22/2022]
|
26
|
Mäki-Marttunen T, Krull F, Bettella F, Hagen E, Næss S, Ness TV, Moberget T, Elvsåshagen T, Metzner C, Devor A, Edwards AG, Fyhn M, Djurovic S, Dale AM, Andreassen OA, Einevoll GT. Alterations in Schizophrenia-Associated Genes Can Lead to Increased Power in Delta Oscillations. Cereb Cortex 2019; 29:875-891. [PMID: 30475994 PMCID: PMC6319172 DOI: 10.1093/cercor/bhy291] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/03/2018] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies have implicated many ion channels in schizophrenia pathophysiology. Although the functions of these channels are relatively well characterized by single-cell studies, the contributions of common variation in these channels to neurophysiological biomarkers and symptoms of schizophrenia remain elusive. Here, using computational modeling, we show that a common biomarker of schizophrenia, namely, an increase in delta-oscillation power, may be a direct consequence of altered expression or kinetics of voltage-gated ion channels or calcium transporters. Our model of a circuit of layer V pyramidal cells highlights multiple types of schizophrenia-related variants that contribute to altered dynamics in the delta-frequency band. Moreover, our model predicts that the same membrane mechanisms that increase the layer V pyramidal cell network gain and response to delta-frequency oscillations may also cause a deficit in a single-cell correlate of the prepulse inhibition, which is a behavioral biomarker highly associated with schizophrenia.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Simula Research Laboratory, Oslo, Norway
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Florian Krull
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Hagen
- Department of Physics, University of Oslo, Oslo, Norway
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Solveig Næss
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Torbjørn V Ness
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Torgeir Moberget
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Christoph Metzner
- Centre for Computer Science and Informatics Research, University of Hertfordshire, Hatfield, UK
| | - Anna Devor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Gaute T Einevoll
- Department of Physics, University of Oslo, Oslo, Norway
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
27
|
Pangrsic T, Singer JH, Koschak A. Voltage-Gated Calcium Channels: Key Players in Sensory Coding in the Retina and the Inner Ear. Physiol Rev 2019; 98:2063-2096. [PMID: 30067155 DOI: 10.1152/physrev.00030.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Calcium influx through voltage-gated Ca (CaV) channels is the first step in synaptic transmission. This review concerns CaV channels at ribbon synapses in primary sense organs and their specialization for efficient coding of stimuli in the physical environment. Specifically, we describe molecular, biochemical, and biophysical properties of the CaV channels in sensory receptor cells of the retina, cochlea, and vestibular apparatus, and we consider how such properties might change over the course of development and contribute to synaptic plasticity. We pay particular attention to factors affecting the spatial arrangement of CaV channels at presynaptic, ribbon-type active zones, because the spatial relationship between CaV channels and release sites has been shown to affect synapse function critically in a number of systems. Finally, we review identified synaptopathies affecting sensory systems and arising from dysfunction of L-type, CaV1.3, and CaV1.4 channels or their protein modulatory elements.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Joshua H Singer
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Alexandra Koschak
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
28
|
Niu J, Dick IE, Yang W, Bamgboye MA, Yue DT, Tomaselli G, Inoue T, Ben-Johny M. Allosteric regulators selectively prevent Ca 2+-feedback of Ca V and Na V channels. eLife 2018; 7:35222. [PMID: 30198845 PMCID: PMC6156082 DOI: 10.7554/elife.35222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/09/2018] [Indexed: 12/31/2022] Open
Abstract
Calmodulin (CaM) serves as a pervasive regulatory subunit of CaV1, CaV2, and NaV1 channels, exploiting a functionally conserved carboxy-tail element to afford dynamic Ca2+-feedback of cellular excitability in neurons and cardiomyocytes. Yet this modularity counters functional adaptability, as global changes in ambient CaM indiscriminately alter its targets. Here, we demonstrate that two structurally unrelated proteins, SH3 and cysteine-rich domain (stac) and fibroblast growth factor homologous factors (fhf) selectively diminish Ca2+/CaM-regulation of CaV1 and NaV1 families, respectively. The two proteins operate on allosteric sites within upstream portions of respective channel carboxy-tails, distinct from the CaM-binding interface. Generalizing this mechanism, insertion of a short RxxK binding motif into CaV1.3 carboxy-tail confers synthetic switching of CaM regulation by Mona SH3 domain. Overall, our findings identify a general class of auxiliary proteins that modify Ca2+/CaM signaling to individual targets allowing spatial and temporal orchestration of feedback, and outline strategies for engineering Ca2+/CaM signaling to individual targets.
Collapse
Affiliation(s)
- Jacqueline Niu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Ivy E Dick
- Department of Physiology, University of Maryland, Baltimore, United States
| | - Wanjun Yang
- Department of Cardiology, Johns Hopkins University, Baltimore, United States
| | | | - David T Yue
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Gordon Tomaselli
- Department of Cardiology, Johns Hopkins University, Baltimore, United States
| | - Takanari Inoue
- Department of Cell Biology, Johns Hopkins University, Baltimore, United States.,Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, United States
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, United States
| |
Collapse
|
29
|
Yang T, Choi JE, Soh D, Tobin K, Joiner ML, Hansen M, Lee A. CaBP1 regulates Ca v1 L-type Ca 2+ channels and their coupling to neurite growth and gene transcription in mouse spiral ganglion neurons. Mol Cell Neurosci 2018; 88:342-352. [PMID: 29548764 DOI: 10.1016/j.mcn.2018.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022] Open
Abstract
CaBP1 is a Ca2+ binding protein that is widely expressed in neurons in the brain, retina, and cochlea. In heterologous expression systems, CaBP1 interacts with and regulates voltage-gated Cav Ca2+ channels but whether this is the case in neurons is unknown. Here, we investigated the cellular functions of CaBP1 in cochlear spiral ganglion neurons (SGNs), which express high levels of CaBP1. Consistent with the role of CaBP1 as a suppressor of Ca2+-dependent inactivation (CDI) of Cav1 (L-type) channels, Cav1 currents underwent greater CDI in SGNs from mice lacking CaBP1 (C-KO) than in wild-type (WT) SGNs. The coupling of Cav1 channels to downstream signaling pathways was also disrupted in C-KO SGNs. Activity-dependent repression of neurite growth was significantly blunted and unresponsive to Cav1 antagonists in C-KO SGNs in contrast to WT SGNs. Moreover, Cav1-mediated Ca2+ signals and phosphorylation of cAMP-response element binding protein were reduced in C-KO SGNs compared to WT SGNs. Our findings establish a role for CaBP1 as an essential regulator of Cav1 channels in SGNs and their coupling to downstream pathways controlling activity-dependent transcription and neurite growth.
Collapse
Affiliation(s)
- Tian Yang
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Ji-Eun Choi
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Daniel Soh
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin Tobin
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Mei-Ling Joiner
- Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan Hansen
- Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Lee
- Departments of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Otolaryngology Head-Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Neurology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
30
|
Chemin J, Taiakina V, Monteil A, Piazza M, Guan W, Stephens RF, Kitmitto A, Pang ZP, Dolphin AC, Perez-Reyes E, Dieckmann T, Guillemette JG, Spafford JD. Calmodulin regulates Ca v3 T-type channels at their gating brake. J Biol Chem 2017; 292:20010-20031. [PMID: 28972185 PMCID: PMC5723990 DOI: 10.1074/jbc.m117.807925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
Calcium (Cav1 and Cav2) and sodium channels possess homologous CaM-binding motifs, known as IQ motifs in their C termini, which associate with calmodulin (CaM), a universal calcium sensor. Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif. We illustrate that T-type channels associate with CaM using co-immunoprecipitation experiments and single particle cryo-electron microscopy. We demonstrate that protostome invertebrate (LCav3) and human Cav3.1, Cav3.2, and Cav3.3 T-type channels specifically associate with CaM at helix 2 of the gating brake in the I-II linker of the channels. Isothermal titration calorimetry results revealed that the gating brake and CaM bind each other with high-nanomolar affinity. We show that the gating brake assumes a helical conformation upon binding CaM, with associated conformational changes to both CaM lobes as indicated by amide chemical shifts of the amino acids of CaM in 1H-15N HSQC NMR spectra. Intact Ca2+-binding sites on CaM and an intact gating brake sequence (first 39 amino acids of the I-II linker) were required in Cav3.2 channels to prevent the runaway gating phenotype, a hyperpolarizing shift in voltage sensitivities and faster gating kinetics. We conclude that the presence of high-nanomolar affinity binding sites for CaM at its universal gating brake and its unique form of regulation via the tuning of the voltage range of activity could influence the participation of Cav3 T-type channels in heart and brain rhythms. Our findings may have implications for arrhythmia disorders arising from mutations in the gating brake or CaM.
Collapse
Affiliation(s)
- Jean Chemin
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | | | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | - Michael Piazza
- Departments of Chemistry, Waterloo, Ontario N2L 3G1, Canada
| | - Wendy Guan
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | | | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | | | | | - J David Spafford
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada.
| |
Collapse
|
31
|
Direct visualization of interaction between calmodulin and connexin45. Biochem J 2017; 474:4035-4051. [PMID: 28963343 DOI: 10.1042/bcj20170426] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/12/2017] [Accepted: 09/25/2017] [Indexed: 01/21/2023]
Abstract
Calmodulin (CaM) is an intracellular Ca2+ transducer involved in numerous activities in a broad Ca2+ signaling network. Previous studies have suggested that the Ca2+/CaM complex may participate in gap junction regulation via interaction with putative CaM-binding motifs in connexins; however, evidence of direct interactions between CaM and connexins has remained elusive to date due to challenges related to the study of membrane proteins. Here, we report the first direct interaction of CaM with Cx45 (connexin45) of γ-family in living cells under physiological conditions by monitoring bioluminescence resonance energy transfer. The interaction between CaM and Cx45 in cells is strongly dependent on intracellular Ca2+ concentration and can be blocked by the CaM inhibitor, N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride (W7). We further reveal a CaM-binding site at the cytosolic loop (residues 164-186) of Cx45 using a peptide model. The strong binding (Kd ∼ 5 nM) observed between CaM and Cx45 peptide, monitored by fluorescence-labeled CaM, is found to be Ca2+-dependent. Furthermore, high-resolution nuclear magnetic resonance spectroscopy reveals that CaM and Cx45 peptide binding leads to global chemical shift changes of 15N-labeled CaM, but does not alter the size of the structure. Observations involving both N- and C-domains of CaM to interact with the Cx45 peptide differ from the embraced interaction with Cx50 from another connexin family. Such interaction further increases Ca2+ sensitivity of CaM, especially at the N-terminal domain. Results of the present study suggest that both helicity and the interaction mode of the cytosolic loop are likely to contribute to CaM's modulation of connexins.
Collapse
|
32
|
Mäki-Marttunen T, Lines GT, Edwards AG, Tveito A, Dale AM, Einevoll GT, Andreassen OA. Pleiotropic effects of schizophrenia-associated genetic variants in neuron firing and cardiac pacemaking revealed by computational modeling. Transl Psychiatry 2017; 7:5. [PMID: 30446648 PMCID: PMC5802468 DOI: 10.1038/s41398-017-0007-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 01/01/2023] Open
Abstract
Schizophrenia patients have an increased risk of cardiac dysfunction. A possible factor underlying this comorbidity are the common variants in the large set of genes that have recently been discovered in genome-wide association studies (GWASs) as risk genes of schizophrenia. Many of these genes control the cell electrogenesis and calcium homeostasis. We applied biophysically detailed models of layer V pyramidal cells and sinoatrial node cells to study the contribution of schizophrenia-associated genes on cellular excitability. By including data from functional genomics literature to simulate the effects of common variants of these genes, we showed that variants of voltage-gated Na+ channel or hyperpolarization-activated cation channel-encoding genes cause qualitatively similar effects on layer V pyramidal cell and sinoatrial node cell excitability. By contrast, variants of Ca2+ channel or transporter-encoding genes mostly have opposite effects on cellular excitability in the two cell types. We also show that the variants may crucially affect the propagation of the cardiac action potential in the sinus node. These results may help explain some of the cardiac comorbidity in schizophrenia, and may facilitate generation of effective antipsychotic medications without cardiac side-effects such as arrhythmia.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway.
| | - Glenn T. Lines
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Aslak Tveito
- Simula Research Laboratory and Center for Cardiological Innovation, Oslo, Norway
| | - Anders M. Dale
- 0000 0001 2107 4242grid.266100.3Multimodal Imaging Laboratory, UC San Diego, La Jolla, CA USA ,0000 0001 2107 4242grid.266100.3Department of Neurosciences, University of California San Diego, La Jolla, CA USA ,0000 0001 2107 4242grid.266100.3Department of Radiology, University of California, San Diego, La Jolla, CA USA
| | - Gaute T. Einevoll
- 0000 0004 0607 975Xgrid.19477.3cDepartment of Mathematical Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway ,0000 0004 1936 8921grid.5510.1Department of Physics, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- 0000 0004 1936 8921grid.5510.1NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
33
|
Pinggera A, Mackenroth L, Rump A, Schallner J, Beleggia F, Wollnik B, Striessnig J. New gain-of-function mutation shows CACNA1D as recurrently mutated gene in autism spectrum disorders and epilepsy. Hum Mol Genet 2017; 26:2923-2932. [PMID: 28472301 PMCID: PMC5886262 DOI: 10.1093/hmg/ddx175] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 11/12/2022] Open
Abstract
CACNA1D encodes the pore-forming α1-subunit of Cav1.3, an L-type voltage-gated Ca2+-channel. Despite the recent discovery of two de novo missense gain-of-function mutations in Cav1.3 in two individuals with autism spectrum disorder (ASD) and intellectual disability CACNA1D has not been considered a prominent ASD-risk gene in large scale genetic analyses, since such studies primarily focus on likely-disruptive genetic variants. Here we report the discovery and characterization of a third de novo missense mutation in CACNA1D (V401L) in a patient with ASD and epilepsy. For the functional characterization we introduced mutation V401L into two major C-terminal long and short Cav1.3 splice variants, expressed wild-type or mutant channel complexes in tsA-201 cells and performed whole-cell patch-clamp recordings. Mutation V401L, localized within the channel's activation gate, significantly enhanced current densities, shifted voltage dependence of activation and inactivation to more negative voltages and reduced channel inactivation in both Cav1.3 splice variants. Altogether, these gating changes are expected to result in enhanced Ca2+-influx through the channel, thus representing a strong gain-of-function phenotype. Additionally, we also found that mutant channels retained full sensitivity towards the clinically available Ca2+ -channel blocker isradipine. Our findings strengthen the evidence for CACNA1D as a novel candidate autism risk gene and encourage experimental therapy with available channel-blockers for this mutation. The additional presence of seizures and neurological abnormalities in our patient define a novel phenotype partially overlapping with symptoms in two individuals with PASNA (congenital primary aldosteronism, seizures and neurological abnormalities) caused by similar Cav1.3 gain-of-function mutations.
Collapse
Affiliation(s)
- Alexandra Pinggera
- Department of Pharmacology and Toxicology Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | - Jens Schallner
- Abteilung Neuropädiatrie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Filippo Beleggia
- Department I of Internal Medicine, University Hospital of Cologne, 50923 Cologne, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
34
|
Ortner NJ, Bock G, Dougalis A, Kharitonova M, Duda J, Hess S, Tuluc P, Pomberger T, Stefanova N, Pitterl F, Ciossek T, Oberacher H, Draheim HJ, Kloppenburg P, Liss B, Striessnig J. Lower Affinity of Isradipine for L-Type Ca 2+ Channels during Substantia Nigra Dopamine Neuron-Like Activity: Implications for Neuroprotection in Parkinson's Disease. J Neurosci 2017; 37:6761-6777. [PMID: 28592699 PMCID: PMC6596555 DOI: 10.1523/jneurosci.2946-16.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 05/08/2017] [Accepted: 05/13/2017] [Indexed: 12/21/2022] Open
Abstract
Ca2+-influx through L-type Ca2+-channels (LTCCs) is associated with activity-related stressful oscillations of Ca2+ levels within dopaminergic (DA) neurons in the substantia nigra (SN), which may contribute to their selective degeneration in Parkinson's disease (PD). LTCC blockers were neuroprotective in mouse neurotoxin models of PD, and isradipine is currently undergoing testing in a phase III clinical trial in early PD. We report no evidence for neuroprotection by in vivo pretreatment with therapeutically relevant isradipine plasma levels, or Cav1.3 LTCC deficiency in 6-OHDA-treated male mice. To explain this finding, we investigated the pharmacological properties of human LTCCs during SN DA-like and arterial smooth muscle (aSM)-like activity patterns using whole-cell patch-clamp recordings in HEK293 cells (Cav1.2 α1-subunit, long and short Cav1.3 α1-subunit splice variants; β3/α2δ1). During SN DA-like pacemaking, only Cav1.3 variants conducted Ca2+ current (ICa) at subthreshold potentials between action potentials. SN DA-like burst activity increased integrated ICa during (Cav1.2 plus Cav1.3) and after (Cav1.3) the burst. Isradipine inhibition was splice variant and isoform dependent, with a 5- to 11-fold lower sensitivity to Cav1.3 variants during SN DA-like pacemaking compared with Cav1.2 during aSM-like activity. Supratherapeutic isradipine concentrations reduced the pacemaker precision of adult mouse SN DA neurons but did not affect their somatic Ca2+ oscillations. Our data predict that Cav1.2 and Cav1.3 splice variants contribute differentially to Ca2+ load in SN DA neurons, with prominent Cav1.3-mediated ICa between action potentials and after bursts. The failure of therapeutically relevant isradipine levels to protect SN DA neurons can be explained by weaker state-dependent inhibition of SN DA LTCCs compared with aSM Cav1.2.SIGNIFICANCE STATEMENT The high vulnerability of dopamine (DA) neurons in the substantia nigra (SN) to neurodegenerative stressors causes Parkinson's disease (PD). Ca2+ influx through voltage-gated L-type Ca2+ channels (LTCCs), in particular Cav1.3, appears to contribute to this vulnerability, and the LTCC inhibitor isradipine is currently being tested as a neuroprotective agent for PD in a phase III clinical trial. However, in our study isradipine plasma concentrations approved for therapy were not neuroprotective in a PD mouse model. We provide an explanation for this observation by demonstrating that during SN DA-like neuronal activity LTCCs are less sensitive to isradipine than Cav1.2 LTCCs in resistance blood vessels (mediating dose-limiting vasodilating effects) and even at supratherapeutic concentrations isradipine fails to reduce somatic Ca2+ oscillations of SN DA neurons.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Gabriella Bock
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Antonios Dougalis
- Institute of Applied Physiology, University of Ulm, 89081 Ulm, Germany
| | - Maria Kharitonova
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Johanna Duda
- Institute of Applied Physiology, University of Ulm, 89081 Ulm, Germany
| | - Simon Hess
- Biocenter, Institute for Zoology, and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50674 Cologne, Germany
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Pomberger
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Florian Pitterl
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Ciossek
- Boehringer Ingelheim Pharma GmbH & Co KG, CNS Research, 88400 Biberach an der Riss, Germany, and
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Henning J Draheim
- Boehringer Ingelheim Pharma GmbH & Co KG, CNS Research, 88400 Biberach an der Riss, Germany, and
| | - Peter Kloppenburg
- Biocenter, Institute for Zoology, and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50674 Cologne, Germany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, 89081 Ulm, Germany
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria,
| |
Collapse
|
35
|
Grassmeyer JJ, Thoreson WB. Synaptic Ribbon Active Zones in Cone Photoreceptors Operate Independently from One Another. Front Cell Neurosci 2017; 11:198. [PMID: 28744203 PMCID: PMC5504102 DOI: 10.3389/fncel.2017.00198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/26/2017] [Indexed: 12/04/2022] Open
Abstract
Cone photoreceptors depolarize in darkness to release glutamate-laden synaptic vesicles. Essential to release is the synaptic ribbon, a structure that helps organize active zones by clustering vesicles near proteins that mediate exocytosis, including voltage-gated Ca2+ channels. Cone terminals have many ribbon-style active zones at which second-order neurons receive input. We asked whether there are functionally significant differences in local Ca2+ influx among ribbons in individual cones. We combined confocal Ca2+ imaging to measure Ca2+ influx at individual ribbons and patch clamp recordings to record whole-cell ICa in salamander cones. We found that the voltage for half-maximal activation (V50) of whole cell ICa in cones averaged −38.1 mV ± 3.05 mV (standard deviation [SD]), close to the cone membrane potential in darkness of ca. −40 mV. Ca2+ signals at individual ribbons varied in amplitude from one another and showed greater variability in V50 values than whole-cell ICa, suggesting that Ca2+ signals can differ significantly among ribbons within cones. After accounting for potential sources of technical variability in measurements of Ca2+ signals and for contributions from cone-to-cone differences in ICa, we found that the variability in V50 values for ribbon Ca2+ signals within individual cones showed a SD of 2.5 mV. Simulating local differences in Ca2+ channel activity at two ribbons by shifting the V50 value of ICa by ±2.5 mV (1 SD) about the mean suggests that when the membrane depolarizes to −40 mV, two ribbons could experience differences in Ca2+ influx of >45%. Further evidence that local Ca2+ changes at ribbons can be regulated independently was obtained in experiments showing that activation of inhibitory feedback from horizontal cells (HCs) to cones in paired recordings changed both amplitude and V50 of Ca2+ signals at individual ribbons. By varying the strength of synaptic output, differences in voltage dependence and amplitude of Ca2+ signals at individual ribbons shape the information transmitted from cones to downstream neurons in vision.
Collapse
Affiliation(s)
- Justin J Grassmeyer
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical CenterOmaha, NE, United States.,Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical CenterOmaha, NE, United States
| | - Wallace B Thoreson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical CenterOmaha, NE, United States.,Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical CenterOmaha, NE, United States
| |
Collapse
|
36
|
Picher MM, Oprişoreanu AM, Jung S, Michel K, Schoch S, Moser T. Rab Interacting Molecules 2 and 3 Directly Interact with the Pore-Forming Ca V1.3 Ca 2+ Channel Subunit and Promote Its Membrane Expression. Front Cell Neurosci 2017. [PMID: 28642685 PMCID: PMC5462952 DOI: 10.3389/fncel.2017.00160] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rab interacting molecules (RIMs) are multi-domain proteins that positively regulate the number of Ca2+ channels at the presynaptic active zone (AZ). Several molecular mechanisms have been demonstrated for RIM-binding to components of the presynaptic Ca2+ channel complex, the key signaling element at the AZ. Here, we report an interaction of the C2B domain of RIM2α and RIM3γ with the C-terminus of the pore-forming α-subunit of CaV1.3 channels (CaV1.3α1), which mediate stimulus-secretion coupling at the ribbon synapses of cochlear inner hair cells (IHCs). Co-expressing full-length RIM2α with a Ca2+ channel complex closely resembling that of IHCs (CaV1.3α1-CaVß2a) in HEK293 cells doubled the Ca2+-current and shifted the voltage-dependence of Ca2+ channel activation by approximately +3 mV. Co-expression of the short RIM isoform RIM3γ increased the CaV1.3α1-CaVß2a-mediated Ca2+-influx in HEK293 cells, but disruption of RIM3γ in mice left Ca2+-influx in IHCs and hearing intact. In conclusion, we propose that RIM2α and RIM3γ directly interact with the C-terminus of the pore-forming subunit of CaV1.3 Ca2+ channels and positively regulate their plasma membrane expression in HEK293 cells.
Collapse
Affiliation(s)
- Maria M Picher
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical ChemistryGöttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of GöttingenGöttingen, Germany
| | - Ana-Maria Oprişoreanu
- Institute of Neuropathology and Department of Epileptology, University of BonnBonn, Germany
| | - SangYong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical ChemistryGöttingen, Germany.,Neuro Modulation and Neuro Circuitry Group, Singapore Bioimaging Consortium (SBIC), Biomedical Sciences InstitutesSingapore, Singapore
| | - Katrin Michel
- Institute of Neuropathology and Department of Epileptology, University of BonnBonn, Germany
| | - Susanne Schoch
- Institute of Neuropathology and Department of Epileptology, University of BonnBonn, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical ChemistryGöttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of GöttingenGöttingen, Germany.,Collaborative Research Center 889, University of GöttingenGöttingen, Germany
| |
Collapse
|
37
|
Different Ca V1.3 Channel Isoforms Control Distinct Components of the Synaptic Vesicle Cycle in Auditory Inner Hair Cells. J Neurosci 2017; 37:2960-2975. [PMID: 28193694 DOI: 10.1523/jneurosci.2374-16.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/27/2017] [Accepted: 02/01/2017] [Indexed: 12/16/2022] Open
Abstract
The mechanisms orchestrating transient and sustained exocytosis in auditory inner hair cells (IHCs) remain largely unknown. These exocytotic responses are believed to mobilize sequentially a readily releasable pool of vesicles (RRP) underneath the synaptic ribbons and a slowly releasable pool of vesicles (SRP) at farther distance from them. They are both governed by Cav1.3 channels and require otoferlin as Ca2+ sensor, but whether they use the same Cav1.3 isoforms is still unknown. Using whole-cell patch-clamp recordings in posthearing mice, we show that only a proportion (∼25%) of the total Ca2+ current in IHCs displaying fast inactivation and resistance to 20 μm nifedipine, a l-type Ca2+ channel blocker, is sufficient to trigger RRP but not SRP exocytosis. This Ca2+ current is likely conducted by short C-terminal isoforms of Cav1.3 channels, notably Cav1.342A and Cav1.343S, because their mRNA is highly expressed in wild-type IHCs but poorly expressed in Otof-/- IHCs, the latter having Ca2+ currents with considerably reduced inactivation. Nifedipine-resistant RRP exocytosis was poorly affected by 5 mm intracellular EGTA, suggesting that the Cav1.3 short isoforms are closely associated with the release site at the synaptic ribbons. Conversely, our results suggest that Cav1.3 long isoforms, which carry ∼75% of the total IHC Ca2+ current with slow inactivation and confer high sensitivity to nifedipine and to internal EGTA, are essentially involved in recruiting SRP vesicles. Intracellular Ca2+ imaging showed that Cav1.3 long isoforms support a deep intracellular diffusion of Ca2+SIGNIFICANCE STATEMENT Auditory inner hair cells (IHCs) encode sounds into nerve impulses through fast and indefatigable Ca2+-dependent exocytosis at their ribbon synapses. We show that this synaptic process involves long and short C-terminal isoforms of the Cav1.3 Ca2+ channel that differ in the kinetics of their Ca2+-dependent inactivation and their relative sensitivity to the l-type Ca2+ channel blocker nifedipine. The short C-terminal isoforms, having fast inactivation and low sensitivity to nifedipine, mainly control the fast fusion of the readily releasable pool (RRP); that is, they encode the phasic exocytotic component. The long isoforms, with slow inactivation and great sensitivity to nifedipine, mainly regulate the vesicular replenishment of the RRP; that is, the sustained or tonic exocytosis.
Collapse
|
38
|
Ca 2+-binding protein 2 inhibits Ca 2+-channel inactivation in mouse inner hair cells. Proc Natl Acad Sci U S A 2017; 114:E1717-E1726. [PMID: 28183797 DOI: 10.1073/pnas.1617533114] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ca2+-binding protein 2 (CaBP2) inhibits the inactivation of heterologously expressed voltage-gated Ca2+ channels of type 1.3 (CaV1.3) and is defective in human autosomal-recessive deafness 93 (DFNB93). Here, we report a newly identified mutation in CABP2 that causes a moderate hearing impairment likely via nonsense-mediated decay of CABP2-mRNA. To study the mechanism of hearing impairment resulting from CABP2 loss of function, we disrupted Cabp2 in mice (Cabp2LacZ/LacZ ). CaBP2 was expressed by cochlear hair cells, preferentially in inner hair cells (IHCs), and was lacking from the postsynaptic spiral ganglion neurons (SGNs). Cabp2LacZ/LacZ mice displayed intact cochlear amplification but impaired auditory brainstem responses. Patch-clamp recordings from Cabp2LacZ/LacZ IHCs revealed enhanced Ca2+-channel inactivation. The voltage dependence of activation and the number of Ca2+ channels appeared normal in Cabp2LacZ/LacZ mice, as were ribbon synapse counts. Recordings from single SGNs showed reduced spontaneous and sound-evoked firing rates. We propose that CaBP2 inhibits CaV1.3 Ca2+-channel inactivation, and thus sustains the availability of CaV1.3 Ca2+ channels for synaptic sound encoding. Therefore, we conclude that human deafness DFNB93 is an auditory synaptopathy.
Collapse
|
39
|
Liu N, Yang Y, Ge L, Liu M, Colecraft HM, Liu X. Cooperative and acute inhibition by multiple C-terminal motifs of L-type Ca 2+ channels. eLife 2017; 6. [PMID: 28059704 PMCID: PMC5279948 DOI: 10.7554/elife.21989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Inhibitions and antagonists of L-type Ca2+ channels are important to both research and therapeutics. Here, we report C-terminus mediated inhibition (CMI) for CaV1.3 that multiple motifs coordinate to tune down Ca2+ current and Ca2+ influx toward the lower limits determined by end-stage CDI (Ca2+-dependent inactivation). Among IQV (preIQ3-IQ domain), PCRD and DCRD (proximal or distal C-terminal regulatory domain), spatial closeness of any two modules, e.g., by constitutive fusion, facilitates the trio to form the complex, compete against calmodulin, and alter the gating. Acute CMI by rapamycin-inducible heterodimerization helps reconcile the concurrent activation/inactivation attenuations to ensure Ca2+ influx is reduced, in that Ca2+ current activated by depolarization is potently (~65%) inhibited at the peak (full activation), but not later on (end-stage inactivation, ~300 ms). Meanwhile, CMI provides a new paradigm to develop CaV1 inhibitors, the therapeutic potential of which is implied by computational modeling of CaV1.3 dysregulations related to Parkinson’s disease. DOI:http://dx.doi.org/10.7554/eLife.21989.001 All cells need calcium ions to stay healthy, but having too many calcium ions can interfere with important processes in the cell and cause severe problems. Proteins known as calcium channels on the cell surface allow calcium ions to flow into the cell from the surrounding environment. Cells carefully control the opening and closing of these channels to prevent too many calcium ions entering the cell at once. CaV1.3 channels are a type of calcium channel that are important for the heart and brain to work properly. Defects in CaV1.3 channels can lead to irregular heart rhythms and neurodegenerative diseases such as Parkinson’s disease. Studies have shown that part of the CaV1.3 channel that sits inside the cell – known as the “tail” – responds to increases in the levels of calcium ions inside the cell by closing the channel. The tail region of CaV1.3 contains three modules, but how these modules work together to regulate channel activity is not clear. Liu, Yang et al. investigated whether the three modules need to be physically connected to each other in the channel protein. For the experiments, several versions of the protein were constructed with different combinations of tail modules being directly linked as part of the same molecule or present as separate molecules. When any two modules were directly linked, the third module could bind to them and this was enough to close the CaV1.3 channel. However, the channel did not close if the modules were totally isolated from each other as three separate molecules. Certain types of neurons in the brain produce electrical signals in a rhythmic fashion that depends on CaV1.3 channels. In Parkinson’s disease, increased movement of calcium ions into these neurons via CaV1.3 channels interferes with the rhythms of the signals and can cause these cells to die. Liu, Yang et al. performed computer simulations to analyse the effects of closing CaV1.3 channels in these neurons. The results suggest that this can restore normal rhythms of electrical activity and prevent these cells from dying. The next step is to understand the molecular details of how the tail region closes CaV1.3 channels and its role in healthy and diseased cells. This may lead to new ways to block CaV1.3 channels in different types of diseases. DOI:http://dx.doi.org/10.7554/eLife.21989.002
Collapse
Affiliation(s)
- Nan Liu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Yaxiong Yang
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Lin Ge
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Min Liu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Xiaodong Liu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| |
Collapse
|
40
|
Waraczynski M, Abbott S, Schultz AV. CaV 1.3 channel blockade in the extended amygdala has a delayed effect on the reward efficacy of medial forebrain bundle stimulation. Behav Brain Res 2016; 317:485-493. [PMID: 27743939 DOI: 10.1016/j.bbr.2016.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 01/27/2023]
Abstract
Previous work in our laboratory has shown that stimulating D2 dopamine receptors in the central sublenticular extended amygdala (SLEAc) can render medial forebrain bundle (MFB) stimulation less rewarding. One of the many ways in which D2 stimulation could affect the activity status of SLEAc neurons is by indirectly blocking calcium ion (Ca2+) influx through CaV1.3 channels. He we directly investigate the effects of blocking CaV1.3 channels on the rewarding effect of MFB stimulation. In experiment one, CaV1.3 blockade with the phenylalkylamine verapamil (2.5 and 5.0μg) or the benzothiazepine diltiazem (5.0 and 10.0μg) did not significantly decrease MFB stimulation's reward efficacy relative to injections of saline. However, there were indications of an unanticipated 24-h-delayed effect of the higher dose of diltiazem - injected ipsilateral to the stimulation site - on the stimulation pulse frequency required to maintain half-maximal response rates ("required frequency"). Experiment two focused on and tracked the time course of this effect. Injections of 10μg of diltiazem decreased required frequency significantly more than did saline injections 24h after injection but not immediately after injection. Required frequency values returned to baseline levels within 48h after injection. This time course is consistent with cellular processes that regulate the insertion of GABA-A receptors in neural membranes. GABA-A-mediated neural communication is implicated in maintaining basal forebrain medium spiny neurons in an excitable state. Therefore, these results may indicate that sustaining SLEAc neurons in an excitable state may be important for MFB stimulation to retain its rewarding properties.
Collapse
Affiliation(s)
- Meg Waraczynski
- Departments of Psychology and Biological Sciences, University of Wisconsin-Whitewater, 800 West, Main Street, Whitewater, WI, USA.
| | - Samantha Abbott
- Departments of Psychology and Biological Sciences, University of Wisconsin-Whitewater, 800 West, Main Street, Whitewater, WI, USA
| | - Alex V Schultz
- Departments of Psychology and Biological Sciences, University of Wisconsin-Whitewater, 800 West, Main Street, Whitewater, WI, USA
| |
Collapse
|
41
|
Surmeier DJ, Schumacker PT, Guzman JD, Ilijic E, Yang B, Zampese E. Calcium and Parkinson's disease. Biochem Biophys Res Commun 2016; 483:1013-1019. [PMID: 27590583 DOI: 10.1016/j.bbrc.2016.08.168] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 01/07/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world. Its causes are poorly understood and there is no proven therapeutic strategy for slowing disease progression. The core motor symptoms of PD are caused by the death of dopaminergic neurons in the substantia nigra pars compacta (SNc). In these neurons, Ca2+entry through plasma membrane Cav1 channels drives a sustained feed-forward stimulation of mitochondrial oxidative phosphorylation. Although this design helps prevent bioenergetic failure when activity needs to be sustained, it leads to basal mitochondrial oxidant stress. Over decades, this basal oxidant stress could compromise mitochondrial function and increase mitophagy, resulting in increased vulnerability to other proteostatic stressors, like elevated alpha synuclein expression. Because this feedforward mechanism is no longer demanded by our lifestyle, it could be dispensed with. Indeed, use of dihydropyridines - negative allosteric modulators of Cav1 Ca2+ channels - comes with little or no effect on brain function but is associated with decreased risk and progression of PD. An ongoing, NIH sponsored, Phase 3 clinical trial in North America is testing the ability of one member of the dihydropyridine class (isradipine) to slow PD progression in early stage patients. The review summarizes the rationale for the trial and outlines some unanswered questions.
Collapse
Affiliation(s)
- D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA.
| | - Paul T Schumacker
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Jaime D Guzman
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Ema Ilijic
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Ben Yang
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| | - Enrico Zampese
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, Illinois, USA
| |
Collapse
|
42
|
Sang L, Dick IE, Yue DT. Protein kinase A modulation of CaV1.4 calcium channels. Nat Commun 2016; 7:12239. [PMID: 27456671 PMCID: PMC4963476 DOI: 10.1038/ncomms12239] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 06/07/2016] [Indexed: 11/19/2022] Open
Abstract
The regulation of L-type Ca2+ channels by protein kinase A (PKA) represents a crucial element within cardiac, skeletal muscle and neurological systems. Although much work has been done to understand this regulation in cardiac CaV1.2 Ca2+ channels, relatively little is known about the closely related CaV1.4 L-type Ca2+ channels, which feature prominently in the visual system. Here we find that CaV1.4 channels are indeed modulated by PKA phosphorylation within the inhibitor of Ca2+-dependent inactivation (ICDI) motif. Phosphorylation of this region promotes the occupancy of calmodulin on the channel, thus increasing channel open probability (PO) and Ca2+-dependent inactivation. Although this interaction seems specific to CaV1.4 channels, introduction of ICDI1.4 to CaV1.3 or CaV1.2 channels endows these channels with a form of PKA modulation, previously unobserved in heterologous systems. Thus, this mechanism may not only play an important role in the visual system but may be generalizable across the L-type channel family. Phosphorylation of L-type calcium CaV channels by protein kinase A is essential for several physiological events. Here, the authors show how this kinase regulates CaV1.4 activity, suggesting a general regulatory mechanism for all L-type calcium channels.
Collapse
Affiliation(s)
- Lingjie Sang
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, Maryland 21205, USA
| | - Ivy E Dick
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, Maryland 21205, USA
| | - David T Yue
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, Maryland 21205, USA.,Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
43
|
Hair cells use active zones with different voltage dependence of Ca2+ influx to decompose sounds into complementary neural codes. Proc Natl Acad Sci U S A 2016; 113:E4716-25. [PMID: 27462107 DOI: 10.1073/pnas.1605737113] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
For sounds of a given frequency, spiral ganglion neurons (SGNs) with different thresholds and dynamic ranges collectively encode the wide range of audible sound pressures. Heterogeneity of synapses between inner hair cells (IHCs) and SGNs is an attractive candidate mechanism for generating complementary neural codes covering the entire dynamic range. Here, we quantified active zone (AZ) properties as a function of AZ position within mouse IHCs by combining patch clamp and imaging of presynaptic Ca(2+) influx and by immunohistochemistry. We report substantial AZ heterogeneity whereby the voltage of half-maximal activation of Ca(2+) influx ranged over ∼20 mV. Ca(2+) influx at AZs facing away from the ganglion activated at weaker depolarizations. Estimates of AZ size and Ca(2+) channel number were correlated and larger when AZs faced the ganglion. Disruption of the deafness gene GIPC3 in mice shifted the activation of presynaptic Ca(2+) influx to more hyperpolarized potentials and increased the spontaneous SGN discharge. Moreover, Gipc3 disruption enhanced Ca(2+) influx and exocytosis in IHCs, reversed the spatial gradient of maximal Ca(2+) influx in IHCs, and increased the maximal firing rate of SGNs at sound onset. We propose that IHCs diversify Ca(2+) channel properties among AZs and thereby contribute to decomposing auditory information into complementary representations in SGNs.
Collapse
|
44
|
Striessnig J, Ortner NJ, Pinggera A. Pharmacology of L-type Calcium Channels: Novel Drugs for Old Targets? Curr Mol Pharmacol 2016; 8:110-22. [PMID: 25966690 PMCID: PMC5384371 DOI: 10.2174/1874467208666150507105845] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/10/2015] [Accepted: 04/20/2015] [Indexed: 11/22/2022]
Abstract
Inhibition of voltage-gated L-type calcium channels by organic calcium channel blockers is a well-established pharmacodynamic concept for the treatment of hypertension and cardiac ischemia. Since decades these antihypertensives (such as the dihydropyridines amlodipine, felodipine or nifedipine) belong to the most widely prescribed drugs
world-wide. Their tolerability is excellent because at therapeutic doses their pharmacological effects in humans are limited to the cardiovascular system. During the last years substantial progress has been made to reveal the physiological role of different L-type calcium channel isoforms in many other tissues, including the brain, endocrine and sensory cells.
Moreover, there is accumulating evidence about their involvement in various human diseases, such as Parkinson's disease, neuropsychiatric disorders and hyperaldosteronism. In this review we discuss the pathogenetic role of L-type calcium channels, potential new indications for existing or isoform-selective compounds and strategies to minimize potential side effects.
Collapse
Affiliation(s)
- Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, A-6020 Innsbruck, Austria.
| | | | | |
Collapse
|
45
|
Ben-Johny M, Dick IE, Sang L, Limpitikul WB, Kang PW, Niu J, Banerjee R, Yang W, Babich JS, Issa JB, Lee SR, Namkung H, Li J, Zhang M, Yang PS, Bazzazi H, Adams PJ, Joshi-Mukherjee R, Yue DN, Yue DT. Towards a Unified Theory of Calmodulin Regulation (Calmodulation) of Voltage-Gated Calcium and Sodium Channels. Curr Mol Pharmacol 2016; 8:188-205. [PMID: 25966688 DOI: 10.2174/1874467208666150507110359] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 01/29/2015] [Accepted: 04/20/2015] [Indexed: 12/13/2022]
Abstract
Voltage-gated Na and Ca(2+) channels represent two major ion channel families that enable myriad biological functions including the generation of action potentials and the coupling of electrical and chemical signaling in cells. Calmodulin regulation (calmodulation) of these ion channels comprises a vital feedback mechanism with distinct physiological implications. Though long-sought, a shared understanding of the channel families remained elusive for two decades as the functional manifestations and the structural underpinnings of this modulation often appeared to diverge. Here, we review recent advancements in the understanding of calmodulation of Ca(2+) and Na channels that suggest a remarkable similarity in their regulatory scheme. This interrelation between the two channel families now paves the way towards a unified mechanistic framework to understand vital calmodulin-dependent feedback and offers shared principles to approach related channelopathic diseases. An exciting era of synergistic study now looms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David T Yue
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
Buchanan PJ, McCloskey KD. Ca V channels and cancer: canonical functions indicate benefits of repurposed drugs as cancer therapeutics. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:621-633. [PMID: 27342111 PMCID: PMC5045480 DOI: 10.1007/s00249-016-1144-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/17/2016] [Accepted: 05/23/2016] [Indexed: 01/19/2023]
Abstract
The importance of ion channels in the hallmarks of many cancers is increasingly recognised. This article reviews current knowledge of the expression of members of the voltage-gated calcium channel family (CaV) in cancer at the gene and protein level and discusses their potential functional roles. The ten members of the CaV channel family are classified according to expression of their pore-forming α-subunit; moreover, co-expression of accessory α2δ, β and γ confers a spectrum of biophysical characteristics including voltage dependence of activation and inactivation, current amplitude and activation/inactivation kinetics. CaV channels have traditionally been studied in excitable cells including neurones, smooth muscle, skeletal muscle and cardiac cells, and drugs targeting the channels are used in the treatment of hypertension and epilepsy. There is emerging evidence that several CaV channels are differentially expressed in cancer cells compared to their normal counterparts. Interestingly, a number of CaV channels also have non-canonical functions and are involved in transcriptional regulation of the expression of other proteins including potassium channels. Pharmacological studies show that CaV canonical function contributes to the fundamental biology of proliferation, cell-cycle progression and apoptosis. This raises the intriguing possibility that calcium channel blockers, approved for the treatment of other conditions, could be repurposed to treat particular cancers. Further research will reveal the full extent of both the canonical and non-canonical functions of CaV channels in cancer and whether calcium channel blockers are beneficial in cancer treatment.
Collapse
Affiliation(s)
- Paul J Buchanan
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7AE, UK.,National Institute of Cellular Biotechnology, School of Nursing and Human Science, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Karen D McCloskey
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7AE, UK.
| |
Collapse
|
47
|
Limpitikul WB, Dick IE, Ben-Johny M, Yue DT. An autism-associated mutation in CaV1.3 channels has opposing effects on voltage- and Ca(2+)-dependent regulation. Sci Rep 2016; 6:27235. [PMID: 27255217 PMCID: PMC4891671 DOI: 10.1038/srep27235] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/13/2016] [Indexed: 01/07/2023] Open
Abstract
CaV1.3 channels are a major class of L-type Ca(2+) channels which contribute to the rhythmicity of the heart and brain. In the brain, these channels are vital for excitation-transcription coupling, synaptic plasticity, and neuronal firing. Moreover, disruption of CaV1.3 function has been associated with several neurological disorders. Here, we focus on the de novo missense mutation A760G which has been linked to autism spectrum disorder (ASD). To explore the role of this mutation in ASD pathogenesis, we examined the effects of A760G on CaV1.3 channel gating and regulation. Introduction of the mutation severely diminished the Ca(2+)-dependent inactivation (CDI) of CaV1.3 channels, an important feedback system required for Ca(2+) homeostasis. This reduction in CDI was observed in two major channel splice variants, though to different extents. Using an allosteric model of channel gating, we found that the underlying mechanism of CDI reduction is likely due to enhanced channel opening within the Ca(2+)-inactivated mode. Remarkably, the A760G mutation also caused an opposite increase in voltage-dependent inactivation (VDI), resulting in a multifaceted mechanism underlying ASD. When combined, these regulatory deficits appear to increase the intracellular Ca(2+) concentration, thus potentially disrupting neuronal development and synapse formation, ultimately leading to ASD.
Collapse
Affiliation(s)
- Worawan B Limpitikul
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713,720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Ivy E Dick
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713,720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Manu Ben-Johny
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713,720 Rutland Avenue, Baltimore, MD 21205, USA
| | - David T Yue
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713,720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
48
|
Kolaj M, Zhang L, Renaud LP. L-type calcium channels and MAP kinase contribute to thyrotropin-releasing hormone-induced depolarization in thalamic paraventricular nucleus neurons. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1120-7. [PMID: 27009047 PMCID: PMC4935505 DOI: 10.1152/ajpregu.00082.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 03/18/2016] [Indexed: 12/11/2022]
Abstract
In rat paraventricular thalamic nucleus (PVT) neurons, activation of thyrotropin-releasing hormone (TRH) receptors enhances neuronal excitability via concurrent decrease in a G protein-coupled inwardly rectifying K (GIRK)-like conductance and opening of a cannabinoid receptor-sensitive transient receptor potential canonical (TRPC)-like conductance. Here, we investigated the calcium (Ca(2+)) contribution to the components of this TRH-induced response. TRH-induced membrane depolarization was reduced in the presence of intracellular BAPTA, also in media containing nominally zero [Ca(2+)]o, suggesting a critical role for both intracellular Ca(2+) release and Ca(2+) influx. TRH-induced inward current was unchanged by T-type Ca(2+) channel blockade, but was decreased by blockade of high-voltage-activated Ca(2+) channels (HVACCs). Both the pharmacologically isolated GIRK-like and the TRPC-like components of the TRH-induced response were decreased by nifedipine and increased by BayK8644, implying Ca(2+) influx via L-type Ca(2+) channels. Only the TRPC-like conductance was reduced by either thapsigargin or dantrolene, suggesting a role for ryanodine receptors and Ca(2+)-induced Ca(2+) release in this component of the TRH-induced response. In pituitary and other cell lines, TRH stimulates MAPK. In PVT neurons, only the GIRK-like component of the TRH-induced current was selectively decreased in the presence of PD98059, a MAPK inhibitor. Collectively, the data imply that TRH-induced depolarization and inward current in PVT neurons involve both a dependency on extracellular Ca(2+) influx via opening of L-type Ca(2+) channels, a sensitivity of a TRPC-like component to intracellular Ca(2+) release via ryanodine channels, and a modulation by MAPK of a GIRK-like conductance component.
Collapse
Affiliation(s)
- Miloslav Kolaj
- Ottawa Hospital Research Institute, Neuroscience Program and University of Ottawa, Department of Medicine, Ottawa, Ontario, Canada
| | - Li Zhang
- Ottawa Hospital Research Institute, Neuroscience Program and University of Ottawa, Department of Medicine, Ottawa, Ontario, Canada
| | - Leo P Renaud
- Ottawa Hospital Research Institute, Neuroscience Program and University of Ottawa, Department of Medicine, Ottawa, Ontario, Canada
| |
Collapse
|
49
|
Moreno CM, Dixon RE, Tajada S, Yuan C, Opitz-Araya X, Binder MD, Santana LF. Ca(2+) entry into neurons is facilitated by cooperative gating of clustered CaV1.3 channels. eLife 2016; 5. [PMID: 27187148 PMCID: PMC4869912 DOI: 10.7554/elife.15744] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/09/2016] [Indexed: 11/13/2022] Open
Abstract
CaV1.3 channels regulate excitability in many neurons. As is the case for all voltage-gated channels, it is widely assumed that individual CaV1.3 channels behave independently with respect to voltage-activation, open probability, and facilitation. Here, we report the results of super-resolution imaging, optogenetic, and electrophysiological measurements that refute this long-held view. We found that the short channel isoform (CaV1.3S), but not the long (CaV1.3L), associates in functional clusters of two or more channels that open cooperatively, facilitating Ca(2+) influx. CaV1.3S channels are coupled via a C-terminus-to-C-terminus interaction that requires binding of the incoming Ca(2+) to calmodulin (CaM) and subsequent binding of CaM to the pre-IQ domain of the channels. Physically-coupled channels facilitate Ca(2+) currents as a consequence of their higher open probabilities, leading to increased firing rates in rat hippocampal neurons. We propose that cooperative gating of CaV1.3S channels represents a mechanism for the regulation of Ca(2+) signaling and electrical activity.
Collapse
Affiliation(s)
- Claudia M Moreno
- Department of Physiology and Membrane Biology, University of California, Davis, United States
| | - Rose E Dixon
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Sendoa Tajada
- Department of Physiology and Membrane Biology, University of California, Davis, United States
| | - Can Yuan
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Ximena Opitz-Araya
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Luis F Santana
- Department of Physiology and Membrane Biology, University of California, Davis, United States
| |
Collapse
|
50
|
Haeseleer F, Williams B, Lee A. Characterization of C-terminal Splice Variants of Cav1.4 Ca2+ Channels in Human Retina. J Biol Chem 2016; 291:15663-73. [PMID: 27226626 DOI: 10.1074/jbc.m116.731737] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated Ca(2+) channels (Cav) undergo extensive alternative splicing that greatly enhances their functional diversity in excitable cells. Here, we characterized novel splice variants of the cytoplasmic C-terminal domain of Cav1.4 Ca(2+) channels that regulate neurotransmitter release in photoreceptors in the retina. These variants lack a portion of exon 45 and/or the entire exon 47 (Cav1.4Δex p45, Cav1.4Δex 47, Cav1.4Δex p45,47) and are expressed in the retina of primates but not mice. Although the electrophysiological properties of Cav1.4Δex p45 are similar to those of full-length channels (Cav1.4FL), skipping of exon 47 dramatically alters Cav1.4 function. Deletion of exon 47 removes part of a C-terminal automodulatory domain (CTM) previously shown to suppress Ca(2+)-dependent inactivation (CDI) and to cause a positive shift in the voltage dependence of channel activation. Exon 47 is crucial for these effects of the CTM because variants lacking this exon show intense CDI and activate at more hyperpolarized voltages than Cav1.4FL The robust CDI of Cav1.4Δex 47 is suppressed by CaBP4, a regulator of Cav1.4 channels in photoreceptors. Although CaBP4 enhances activation of Cav1.4FL, Cav1.4Δex 47 shows similar voltage-dependent activation in the presence and absence of CaBP4. We conclude that exon 47 encodes structural determinants that regulate CDI and voltage-dependent activation of Cav1.4, and is necessary for modulation of channel activation by CaBP4.
Collapse
Affiliation(s)
- Françoise Haeseleer
- From the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195 and
| | - Brittany Williams
- the Departments of Molecular Physiology and Biophysics, Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa 52242
| | - Amy Lee
- the Departments of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, Neurology, and
| |
Collapse
|