1
|
Del Rivero Morfin PJ, Chavez DS, Jayaraman S, Yang L, Kochiss AL, Colecraft HM, Liu XS, Marx SO, Rajadhyaksha AM, Ben-Johny M. A Genetically Encoded Actuator Selectively Boosts L-type Calcium Channels in Diverse Physiological Settings. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558856. [PMID: 37790372 PMCID: PMC10542531 DOI: 10.1101/2023.09.22.558856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
L-type Ca 2+ channels (Ca V 1.2/1.3) convey influx of calcium ions (Ca 2+ ) that orchestrate a bevy of biological responses including muscle contraction and gene transcription. Deficits in Ca V 1 function play a vital role in cardiac and neurodevelopmental disorders. Yet conventional pharmacological approaches to upregulate Ca V 1 are limited, as excessive Ca 2+ influx leads to cytotoxicity. Here, we develop a genetically encoded enhancer of Ca V 1.2/1.3 channels (GeeC) to manipulate Ca 2+ entry in distinct physiological settings. Specifically, we functionalized a nanobody that targets the Ca V macromolecular complex by attaching a minimal effector domain from a Ca V enhancer-leucine rich repeat containing protein 10 (Lrrc10). In cardiomyocytes, GeeC evoked a 3-fold increase in L-type current amplitude. In neurons, GeeC augmented excitation-transcription (E-T) coupling. In all, GeeC represents a powerful strategy to boost Ca V 1.2/1.3 function in distinct physiological settings and, in so doing, lays the groundwork to illuminate new insights on neuronal and cardiac physiology and disease.
Collapse
|
2
|
Park JF, Liang J, Umar S. Electrical Remodeling in Right Ventricular Failure Due to Pulmonary Hypertension: Unraveling Novel Therapeutic Targets. Int J Mol Sci 2023; 24:4633. [PMID: 36902065 PMCID: PMC10003421 DOI: 10.3390/ijms24054633] [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: 01/21/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/05/2023] Open
Abstract
Arrhythmias in the setting of right-ventricular (RV) remodeling contribute to majority of deaths in patients with pulmonary hypertension. However, the underlying mechanism of electrical remodeling remains elusive, especially ventricular arrhythmias. Here, we analyzed the RV transcriptome of pulmonary arterial hypertension (PAH) patients with compensated RV or decompensated RV and identified 8 and 45 differentially expressed genes known to be involved in regulating the electrophysiological properties of excitation and contraction of cardiac myocytes, respectively. Transcripts encoding voltage-gated Ca2+ and Na+ channels were notably decreased in PAH patients with decompensated RV, along with significant dysregulation of KV and Kir channels. We further showed similarity of the RV channelome signature with two well-known animal models of PAH, monocrotaline (MCT)- and Sugen-hypoxia (SuHx)-treated rats. We identified 15 common transcripts among MCT, SuHx, and PAH patients with decompensated RV failure. In addition, data-driven drug repurposing using the channelome signature of PAH patients with decompensated RV failure predicted drug candidates that may reverse the altered gene expression. Comparative analysis provided further insight into clinical relevance and potential preclinical therapeutic studies targeting mechanisms involved in arrhythmogenesis.
Collapse
Affiliation(s)
| | | | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Subbamanda YD, Bhargava A. Intercommunication between Voltage-Gated Calcium Channels and Estrogen Receptor/Estrogen Signaling: Insights into Physiological and Pathological Conditions. Cells 2022; 11:cells11233850. [PMID: 36497108 PMCID: PMC9739980 DOI: 10.3390/cells11233850] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Voltage-gated calcium channels (VGCCs) and estrogen receptors are important cellular proteins that have been shown to interact with each other across varied cells and tissues. Estrogen hormone, the ligand for estrogen receptors, can also exert its effects independent of estrogen receptors that collectively constitute non-genomic mechanisms. Here, we provide insights into the VGCC regulation by estrogen and the possible mechanisms involved therein across several cell types. Notably, most of the interaction is described in neuronal and cardiovascular tissues given the importance of VGCCs in these electrically excitable tissues. We describe the modulation of various VGCCs by estrogen known so far in physiological conditions and pathological conditions. We observed that in most in vitro studies higher concentrations of estrogen were used while a handful of in vivo studies used meager concentrations resulting in inhibition or upregulation of VGCCs, respectively. There is a need for more relevant physiological assays to study the regulation of VGCCs by estrogen. Additionally, other interacting receptors and partners need to be identified that may be involved in exerting estrogen receptor-independent effects of estrogen.
Collapse
|
4
|
Bamgboye MA, Herold KG, Vieira DC, Traficante MK, Rogers PJ, Ben-Johny M, Dick IE. CaV1.2 channelopathic mutations evoke diverse pathophysiological mechanisms. J Gen Physiol 2022; 154:e202213209. [PMID: 36167061 PMCID: PMC9524202 DOI: 10.1085/jgp.202213209] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/09/2022] [Accepted: 09/13/2022] [Indexed: 01/17/2023] Open
Abstract
The first pathogenic mutation in CaV1.2 was identified in 2004 and was shown to cause a severe multisystem disorder known as Timothy syndrome (TS). The mutation was localized to the distal S6 region of the channel, a region known to play a major role in channel activation. TS patients suffer from life-threatening cardiac symptoms as well as significant neurodevelopmental deficits, including autism spectrum disorder (ASD). Since this discovery, the number and variety of mutations identified in CaV1.2 have grown tremendously, and the distal S6 regions remain a frequent locus for many of these mutations. While the majority of patients harboring these mutations exhibit cardiac symptoms that can be well explained by known pathogenic mechanisms, the same cannot be said for the ASD or neurodevelopmental phenotypes seen in some patients, indicating a gap in our understanding of the pathogenesis of CaV1.2 channelopathies. Here, we use whole-cell patch clamp, quantitative Ca2+ imaging, and single channel recordings to expand the known mechanisms underlying the pathogenesis of CaV1.2 channelopathies. Specifically, we find that mutations within the S6 region can exert independent and separable effects on activation, voltage-dependent inactivation (VDI), and Ca2+-dependent inactivation (CDI). Moreover, the mechanisms underlying the CDI effects of these mutations are varied and include altered channel opening and possible disruption of CDI transduction. Overall, these results provide a structure-function framework to conceptualize the role of S6 mutations in pathophysiology and offer insight into the biophysical defects associated with distinct clinical manifestations.
Collapse
Affiliation(s)
- Moradeke A. Bamgboye
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Kevin G. Herold
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Daiana C.O. Vieira
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Maria K. Traficante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Philippa J. Rogers
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Manu Ben-Johny
- Department of Physiology and Biophysics, Columbia University, New York, NY
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
5
|
Network-Based Data Analysis Reveals Ion Channel-Related Gene Features in COVID-19: A Bioinformatic Approach. Biochem Genet 2022; 61:471-505. [PMID: 36104591 PMCID: PMC9473477 DOI: 10.1007/s10528-022-10280-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/01/2022] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 (COVID-19) seriously threatens human health and has been disseminated worldwide. Although there are several treatments for COVID-19, its control is currently suboptimal. Therefore, the development of novel strategies to treat COVID-19 is necessary. Ion channels are located on the membranes of all excitable cells and many intracellular organelles and are key components involved in various biological processes. They are a target of interest when searching for drug targets. This study aimed to reveal the relevant molecular features of ion channel genes in COVID-19 based on bioinformatic analyses. The RNA-sequencing data of patients with COVID-19 and healthy subjects (GSE152418 and GSE171110 datasets) were obtained from the Gene Expression Omnibus (GEO) database. Ion channel genes were selected from the Hugo Gene Nomenclature Committee (HGNC) database. The RStudio software was used to process the data based on the corresponding R language package to identify ion channel-associated differentially expressed genes (DEGs). Based on the DEGs, Gene Ontology (GO) functional and pathway enrichment analyses were performed using the Enrichr web tool. The STRING database was used to generate a protein-protein interaction (PPI) network, and the Cytoscape software was used to screen for hub genes in the PPI network based on the cytoHubba plug-in. Transcription factors (TF)-DEG, DEG-microRNA (miRNA) and DEG-disease association networks were constructed using the NetworkAnalyst web tool. Finally, the screened hub genes as drug targets were subjected to enrichment analysis based on the DSigDB using the Enrichr web tool to identify potential therapeutic agents for COVID-19. A total of 29 ion channel-associated DEGs were identified. GO functional analysis showed that the DEGs were integral components of the plasma membrane and were mainly involved in inorganic cation transmembrane transport and ion channel activity functions. Pathway analysis showed that the DEGs were mainly involved in nicotine addiction, calcium regulation in the cardiac cell and neuronal system pathways. The top 10 hub genes screened based on the PPI network included KCNA2, KCNJ4, CACNA1A, CACNA1E, NALCN, KCNA5, CACNA2D1, TRPC1, TRPM3 and KCNN3. The TF-DEG and DEG-miRNA networks revealed significant TFs (FOXC1, GATA2, HINFP, USF2, JUN and NFKB1) and miRNAs (hsa-mir-146a-5p, hsa-mir-27a-3p, hsa-mir-335-5p, hsa-let-7b-5p and hsa-mir-129-2-3p). Gene-disease association network analysis revealed that the DEGs were closely associated with intellectual disability and cerebellar ataxia. Drug-target enrichment analysis showed that the relevant drugs targeting the hub genes CACNA2D1, CACNA1A, CACNA1E, KCNA2 and KCNA5 were gabapentin, gabapentin enacarbil, pregabalin, guanidine hydrochloride and 4-aminopyridine. The results of this study provide a valuable basis for exploring the mechanisms of ion channel genes in COVID-19 and clues for developing therapeutic strategies for COVID-19.
Collapse
|
6
|
Archana GM, Arunkumar RC, Omkumar RV. Assays for L-type voltage gated calcium channels. Anal Biochem 2022; 656:114827. [PMID: 35964733 DOI: 10.1016/j.ab.2022.114827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/10/2022] [Accepted: 07/19/2022] [Indexed: 11/01/2022]
Abstract
Voltage gated calcium channels (VGCCs) are pursued as drug targets for neurodegenerative and cardiovascular diseases. High throughput drug screening targeting VGCCs depends on patch-clamp electrophysiology or fluorophore-based calcium imaging that requires powerful equipment and specialized expertise thus leading to cost escalation. Moreover, VGCC needs to be transfected into cell lines such as HEK-293. We report the presence of L-type VGCC (L-VGCC) subunit proteins, Cav1.2, α2δ and β in HEK-293 cells and the application of simple methods for its assay. Endogenous expression of the channel in HEK-293 cells overcomes the need for transfection. L-VGCC in HEK-293 cells was activated either by the agonist, BayK8644 or by KCl-mediated depolarization. Activity was detected using the calcium sensing probe, GCaMP6m by live imaging. L-VGCC activity induced enhancement in GCaMP6m fluorescence returned to baseline corresponding to channel-closure. Activity was also shown using a methodology involving end-point detection of the calcium dependent interaction of α-CaMKII with NMDA receptor subunit GluN2B sequence. This methodology further simplifies the assay as it eliminates the need for real time imaging. Activation was blocked by the specific L-type VGCC antagonist, nifedipine. Finding the protein and activity of L-VGCC in HEK-293 cells offers commercially viable assays for drug screening.
Collapse
Affiliation(s)
- G M Archana
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P. O., Thiruvananthapuram, 695014, India; University of Kerala, India
| | - R C Arunkumar
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P. O., Thiruvananthapuram, 695014, India; University of Kerala, India
| | - R V Omkumar
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P. O., Thiruvananthapuram, 695014, India.
| |
Collapse
|
7
|
Giang N, Mars M, Moreau M, Mejia JE, Bouchaud G, Magnan A, Michelet M, Ronsin B, Murphy GG, Striessnig J, Guéry J, Pelletier L, Savignac M. Separation of the Ca V 1.2-Ca V 1.3 calcium channel duo prevents type 2 allergic airway inflammation. Allergy 2022; 77:525-539. [PMID: 34181765 DOI: 10.1111/all.14993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/16/2021] [Accepted: 05/16/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Voltage-gated calcium (Cav 1) channels contribute to T-lymphocyte activation. Cav 1.2 and Cav 1.3 channels are expressed in Th2 cells but their respective roles are unknown, which is investigated herein. METHODS We generated mice deleted for Cav 1.2 in T cells or Cav 1.3 and analyzed TCR-driven signaling. In this line, we developed original fast calcium imaging to measure early elementary calcium events (ECE). We also tested the impact of Cav 1.2 or Cav 1.3 deletion in models of type 2 airway inflammation. Finally, we checked whether the expression of both Cav 1.2 and Cav 1.3 in T cells from asthmatic children correlates with Th2-cytokine expression. RESULTS We demonstrated non-redundant and synergistic functions of Cav 1.2 and Cav 1.3 in Th2 cells. Indeed, the deficiency of only one channel in Th2 cells triggers TCR-driven hyporesponsiveness with weakened tyrosine phosphorylation profile, a strong decrease in initial ECE and subsequent reduction in the global calcium response. Moreover, Cav 1.3 has a particular role in calcium homeostasis. In accordance with the singular roles of Cav 1.2 and Cav 1.3 in Th2 cells, deficiency in either one of these channels was sufficient to inhibit cardinal features of type 2 airway inflammation. Furthermore, Cav 1.2 and Cav 1.3 must be co-expressed within the same CD4+ T cell to trigger allergic airway inflammation. Accordingly with the concerted roles of Cav 1.2 and Cav 1.3, the expression of both channels by activated CD4+ T cells from asthmatic children was associated with increased Th2-cytokine transcription. CONCLUSIONS Thus, Cav 1.2 and Cav 1.3 act as a duo, and targeting only one of these channels would be efficient in allergy treatment.
Collapse
Affiliation(s)
- Nicolas Giang
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Marion Mars
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Marc Moreau
- Centre de Biologie du Développement Centre de Biologie Intégrative Université de ToulouseCNRSUniversité Paul Sabatier III Toulouse France
| | - Jose E. Mejia
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | | | - Antoine Magnan
- Institut du Thorax INSERM CNRSUniversité de Nantes Nantes France
- Service de Pneumologie Centre Hospitalier Universitaire de Nantes Nantes France
| | - Marine Michelet
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
- Pediatric Pneumology and Allergology Unit Hôpital des EnfantsCentre Hospitalier Universitaire Toulouse Toulouse France
- Unité de Recherche Clinique Pédiatrique/module plurithématique pédiatrique du CIC Toulouse France
| | - Brice Ronsin
- Centre de Biologie du Développement Centre de Biologie Intégrative Université de ToulouseCNRSUniversité Paul Sabatier III Toulouse France
| | - Geoffrey G. Murphy
- Molecular and Behavioral Neuroscience Institute University of Michigan Ann Arbor MI USA
| | - Joerg Striessnig
- Department of Pharmacology and Toxicology Institute of Pharmacy Center for Molecular Biosciences University of Innsbruck Innsbruck Austria
| | - Jean‐Charles Guéry
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Lucette Pelletier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| | - Magali Savignac
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291 CNRS UMR5051Université Paul Sabatier Toulouse III Toulouse France
| |
Collapse
|
8
|
Yu W. Reviving Cav1.2 as an attractive drug target to treat bladder dysfunction. FASEB J 2022; 36:e22118. [PMID: 34939692 PMCID: PMC9841550 DOI: 10.1096/fj.202101475r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 01/18/2023]
Abstract
Inhibition of bladder contraction with antimuscarinics is a common approach to treat bladder hyperactivity, and the L-type voltage-gated calcium channel α1C (Cav1.2) is crucial for bladder contractility. Therefore, strategies aimed at inhibiting Cav1.2 appear warranted. However, multiple clinical trials that attempted to treat bladder overactivity with calcium channel blockers (CCBs) have been unsuccessful, creating an unsolved mystery. In contrast, cardiologists and epidemiologists have reported strong associations between CCB use and bladder hyperactivity, opposing expectations of urologists. Recent findings from our lab offer a potential explanation. We have demonstrated that ketamine which can cause cystitis, functions, like nifedipine, as a Cav1.2 antagonist. We also show that a Cav1.2 agonist which potentiates muscle contraction, rather than antagonizing it, can increase the volume of voids and reduce voiding frequency. This perspective will discuss in detail the unsuccessful urological trials of CCBs and the promise of Cav1.2 agonists as potential novel therapies for bladder dysfunctions.
Collapse
Affiliation(s)
- Weiqun Yu
- Department of Medicine Beth Israel Deaconess Medical Center and Harvard Medical School Boston Massachuesetts USA
| |
Collapse
|
9
|
Pelletier L, Moreau M. Ca v1 channels is also a story of non excitable cells: Application to calcium signalling in two different non related models. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118996. [PMID: 33675852 DOI: 10.1016/j.bbamcr.2021.118996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
Calcium is a second messenger essential, in all cells, for most cell functions. The spatio-temporal control of changes in intracellular calcium concentration is partly due to the activation of calcium channels. Voltage-operated calcium channels are present in excitable and non-excitable cells. If the mechanism of voltage-operated calcium channels is well known in excitable cells the Ca2+ toolkit used in non-excitable cells to activate the calcium channels is less described. Herein we discuss about very similar pathways involving voltage activated Cav1 channels in two unrelated non-excitable cells; ectoderm cells undergoing neural development and effector Th2 lymphocytes responsible for parasite elimination and also allergic diseases. We will examine the way by which these channels operate and are regulated, as well as the consequences in terms of gene transcription. Finally, we will consider the questions that remain unsolved and how they might be a challenge for the future.
Collapse
Affiliation(s)
- Lucette Pelletier
- Infinity - Toulouse Institute For Infectious and Inflammatory Diseases INSERM UMR1291, CNRS UMR5051, University Toulouse III CHU Purpan, BP 3028, 31024 Toulouse CEDEX 3, France
| | - Marc Moreau
- Université Toulouse3, Centre de biologie du développement, CNRS UMR5547, 118 route de Narbonne, F31062 Toulouse Cedex 04, France.
| |
Collapse
|
10
|
Abstract
This chapter describes what a channelopathy is and how mutations in the genes result in different types of clinical abnormalities. It provides a description of common types of cardiac channelopathies with examples of how there are some areas of overlap with sensory-neuromuscular channelopathies. We describe the cardiac channelopathies of Jervell and Lange-Nielson syndrome, Andersen-Tawil syndrome, Timothy syndrome, catecholaminergic polymorphic ventricular tachycardia, Brugada syndrome, and sinoatrial node dysfunction and deafness. We also discuss sudden unexpected death in epilepsy and how it could relate to some cardiac channelopathies.
Collapse
|
11
|
Tabatabaee MS, Kerkovius J, Menard F. Design of an Imaging Probe to Monitor Real-Time Redistribution of L-type Voltage-Gated Calcium Channels in Astrocytic Glutamate Signaling. Mol Imaging Biol 2021; 23:407-416. [PMID: 33432518 DOI: 10.1007/s11307-020-01573-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE In the brain, astrocytes are non-excitable cells that undergo rapid morphological changes when stimulated by the excitatory neurotransmitter glutamate. We developed a chemical probe to monitor how glutamate affects the density and distribution of astrocytic L-type voltage-gated calcium channels (LTCC). PROCEDURES The imaging probe FluoBar1 was created from a barbiturate ligand modified with a fluorescent coumarin moiety. The probe selectivity was examined with colocalization analyses of confocal fluorescence imaging in U118-MG and transfected COS-7 cells. Living cells treated with 50 nM FluoBar1 were imaged in real time to reveal changes in density and distribution of astrocytic LTCCs upon exposure to glutamate. RESULTS FluoBar1 was synthesized in ten steps. The selectivity of the probe was demonstrated with immunoblotting and confocal imaging of immunostained cells expressing the CaV1.2 isoform of LTCCs proteins. Applying FluoBar1 to astrocyte model cells U118-MG allowed us to measure a fivefold increase in fluorescence density of LTCCs upon glutamate exposure. CONCLUSIONS Imaging probe FluoBar1 allows the real-time monitoring of LTCCs in living cells, revealing for first time that glutamate causes a rapid increase of LTCC membranar density in astrocyte model cells. FluoBar1 may help tackle previously intractable questions about LTCC dynamics in cellular events.
Collapse
Affiliation(s)
- Mitra Sadat Tabatabaee
- Department of Biochemistry & Molecular Biology, I.K. Barber Faculty of Science, University of British Columbia, Kelowna, BC, Canada
| | - Jeff Kerkovius
- Department of Chemistry, I.K. Barber Faculty of Science, University of British Columbia, Kelowna, BC, Canada
| | - Frederic Menard
- Department of Biochemistry & Molecular Biology, I.K. Barber Faculty of Science, University of British Columbia, Kelowna, BC, Canada. .,Department of Chemistry, I.K. Barber Faculty of Science, University of British Columbia, Kelowna, BC, Canada.
| |
Collapse
|
12
|
Hopp SC. Targeting microglia L-type voltage-dependent calcium channels for the treatment of central nervous system disorders. J Neurosci Res 2021; 99:141-162. [PMID: 31997405 PMCID: PMC9394523 DOI: 10.1002/jnr.24585] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+ ) is a ubiquitous mediator of a multitude of cellular functions in the central nervous system (CNS). Intracellular Ca2+ is tightly regulated by cells, including entry via plasma membrane Ca2+ permeable channels. Of specific interest for this review are L-type voltage-dependent Ca2+ channels (L-VDCCs), due to their pleiotropic role in several CNS disorders. Currently, there are numerous approved drugs that target L-VDCCs, including dihydropyridines. These drugs are safe and effective for the treatment of humans with cardiovascular disease and may also confer neuroprotection. Here, we review the potential of L-VDCCs as a target for the treatment of CNS disorders with a focus on microglia L-VDCCs. Microglia, the resident immune cells of the brain, have attracted recent attention for their emerging inflammatory role in several CNS diseases. Intracellular Ca2+ regulates microglia transition from a resting quiescent state to an "activated" immune-effector state and is thus a valuable target for manipulation of microglia phenotype. We will review the literature on L-VDCC expression and function in the CNS and on microglia in vitro and in vivo and explore the therapeutic landscape of L-VDCC-targeting agents at present and future challenges in the context of Alzheimer's disease, Parkinson's disease, Huntington's disease, neuropsychiatric diseases, and other CNS disorders.
Collapse
Affiliation(s)
- Sarah C. Hopp
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
13
|
Investigation of the Selectivity of L-Type Voltage-Gated Calcium Channels 1.3 for Pyrimidine-2,4,6-Triones Derivatives Based on Molecular Dynamics Simulation. Molecules 2020; 25:molecules25225440. [PMID: 33233858 PMCID: PMC7699898 DOI: 10.3390/molecules25225440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 11/17/2022] Open
Abstract
Human Cav1.3 (hCav1.3) is of great interest as a potential target for Parkinson’s disease. However, common medications like dihydropyridines (DHPs), a kind of classic calcium channel blocker, have poor selectivity to hCav1.3 in clinical treatment, mainly due to being implicated in cardiovascular side-effects mediated by human Cav1.2 (hCav1.2). Recently, pyrimidine-2,4,6-triones (PYTs) have received extensive attention as prominent selective inhibitors to hCav1.3. In this study, we describe the selectivity mechanism of PYTs for hCav1.2 and hCav1.3 based on molecular dynamic simulation methods. Our results reveal that the van der Waals (vdW) interaction was the most important force affecting selectivity. Moreover, the hydrophobic interaction was more conducive to the combination. The highly hydrophobic amino acid residues on hCav1.3, such as V162 (IR1), L303 (IR2), M481 (IR3), and F484 (IR3), provided the greatest contributions in the binding free energy. On the other hand, the substituents of a halogen-substituted aromatic ring, cycloalkyl and norbornyl on PYTs, which are pertinent to the steric hindrance of the compounds, played core roles in the selectivity and affinity for hCav1.3, whereas strong polar substituents needed to be avoided. The findings could provide valuable information for designing more effective and safe medicines for Parkinson’s disease.
Collapse
|
14
|
Endres D, Decher N, Röhr I, Vowinkel K, Domschke K, Komlosi K, Tzschach A, Gläser B, Schiele MA, Runge K, Süß P, Schuchardt F, Nickel K, Stallmeyer B, Rinné S, Schulze-Bahr E, Tebartz van Elst L. New Cav1.2 Channelopathy with High-Functioning Autism, Affective Disorder, Severe Dental Enamel Defects, a Short QT Interval, and a Novel CACNA1C Loss-Of-Function Mutation. Int J Mol Sci 2020; 21:ijms21228611. [PMID: 33203140 PMCID: PMC7696251 DOI: 10.3390/ijms21228611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/04/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Complex neuropsychiatric-cardiac syndromes can be genetically determined. For the first time, the authors present a syndromal form of short QT syndrome in a 34-year-old German male patient with extracardiac features with predominant psychiatric manifestation, namely a severe form of secondary high-functioning autism spectrum disorder (ASD), along with affective and psychotic exacerbations, and severe dental enamel defects (with rapid wearing off his teeth) due to a heterozygous loss-of-function mutation in the CACNA1C gene (NM_000719.6: c.2399A > C; p.Lys800Thr). This mutation was found only once in control databases; the mutated lysine is located in the Cav1.2 calcium channel, is highly conserved during evolution, and is predicted to affect protein function by most pathogenicity prediction algorithms. L-type Cav1.2 calcium channels are widely expressed in the brain and heart. In the case presented, electrophysiological studies revealed a prominent reduction in the current amplitude without changes in the gating behavior of the Cav1.2 channel, most likely due to a trafficking defect. Due to the demonstrated loss of function, the p.Lys800Thr variant was finally classified as pathogenic (ACMG class 4 variant) and is likely to cause a newly described Cav1.2 channelopathy.
Collapse
Affiliation(s)
- Dominique Endres
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.R.); (K.N.); (L.T.v.E.)
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.D.); (M.A.S.)
- Correspondence: ; Tel.: +49-761-270-66360
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior-Philipps-University Marburg, 35037 Marburg, Germany; (N.D.); (I.R.); (K.V.); (S.R.)
| | - Isabell Röhr
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior-Philipps-University Marburg, 35037 Marburg, Germany; (N.D.); (I.R.); (K.V.); (S.R.)
| | - Kirsty Vowinkel
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior-Philipps-University Marburg, 35037 Marburg, Germany; (N.D.); (I.R.); (K.V.); (S.R.)
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.D.); (M.A.S.)
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Katalin Komlosi
- Institute of Human Genetics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (K.K.); (A.T.); (B.G.)
| | - Andreas Tzschach
- Institute of Human Genetics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (K.K.); (A.T.); (B.G.)
| | - Birgitta Gläser
- Institute of Human Genetics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (K.K.); (A.T.); (B.G.)
| | - Miriam A. Schiele
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.D.); (M.A.S.)
| | - Kimon Runge
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.R.); (K.N.); (L.T.v.E.)
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.D.); (M.A.S.)
| | - Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Florian Schuchardt
- Department of Neurology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Kathrin Nickel
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.R.); (K.N.); (L.T.v.E.)
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.D.); (M.A.S.)
| | - Birgit Stallmeyer
- Institute for Genetics of Heart Diseases, Department of Cardiovascular Medicine, University Hospital Münster, 48149 Münster, Germany; (B.S.); (E.S.-B.)
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior-Philipps-University Marburg, 35037 Marburg, Germany; (N.D.); (I.R.); (K.V.); (S.R.)
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases, Department of Cardiovascular Medicine, University Hospital Münster, 48149 Münster, Germany; (B.S.); (E.S.-B.)
| | - Ludger Tebartz van Elst
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.R.); (K.N.); (L.T.v.E.)
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (K.D.); (M.A.S.)
| |
Collapse
|
15
|
Tabatabaee MS, Menard F. L-type Voltage-Gated Calcium Channel Modulators Inhibit Glutamate-Induced Morphology Changes in U118-MG Astrocytoma Cells. Cell Mol Neurobiol 2020; 40:1429-1437. [PMID: 32172458 PMCID: PMC11448807 DOI: 10.1007/s10571-020-00828-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/06/2020] [Indexed: 10/24/2022]
Abstract
The excitatory neurotransmitter glutamate evokes physiological responses within the astrocytic network that lead to fine morphological changes. However, the mechanism by which astrocytes couple glutamate sensing with cellular calcium rise remains unclear. We tested a possible connection between L-type voltage-gated calcium channels (Cav) and glutamate-induced response in U118-MG astrocytoma cells. While astrocytoma cells differ from primary astrocytes, they demonstrate the same response to glutamate. In this study, the extension of U118-MG processes upon glutamate exposure was shown to depend on extracellular calcium entry via L-type Cav's. Drugs known to bind to the pore-forming subunit of Cav's decreased the astrocytic filopodia extension caused by glutamate, and ligands of the α2δ auxiliary subunit inhibited all process growth (e.g., gabapentinoids). The observed phenotypic responses suggest that α2δ is a main contributor to the role of Cavs in glutamate-dependent filopodiagenesis, thereby opening new avenues of research on the role of α2δ in astrocytic neurochemical signaling.
Collapse
Affiliation(s)
- Mitra Sadat Tabatabaee
- Department of Biochemistry & Molecular Biology, I.K. Barber School of Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Frederic Menard
- Department of Biochemistry & Molecular Biology, I.K. Barber School of Sciences, University of British Columbia, Kelowna, BC, Canada.
- Department of Chemistry, I.K. Barber School of Sciences, University of British Columbia, Kelowna, BC, Canada.
| |
Collapse
|
16
|
Ma XE, Liu B, Zhao CX. Modulation of Ca 2+-induced Ca 2+ release by ubiquitin protein ligase E3 component n-recognin UBR3 and 6 in cardiac myocytes. Channels (Austin) 2020; 14:326-335. [PMID: 32988261 PMCID: PMC7757829 DOI: 10.1080/19336950.2020.1824957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Ca2+-induced Ca2+ release (CICR) from sarcoplasmic reticulum is a finely tuned process responsible for cardiac excitation and contraction. The ubiquitin–proteasome system (UPS) as a major degradative system plays a crucial role in the maintenance of Ca2+ homeostasis. The E3 component N-recognin (UBR) subfamily is a part of the UPS; however, the role of UBR in regulating cardiac CICR is unknown. In the present study, we found that among the UBR family, single knockdown of UBR3 or UBR6 significantly elevated the amplitude of sarcoplasmic reticulum Ca2+ release without affecting Ca2+ transient decay time in neonatal rat ventricular myocytes. The protein expression of alpha 1 C subunit of L-type voltage-dependent Ca2+ channel (Cav1.2) was increased after UBR3/6 knockdown, whereas the protein levels of RyR2, SERCA2a, and PLB remained unchanged. In line with the increase in Cav1.2 proteins, the UBR3/6 knockdown enhanced the current of Cav1.2 channels. Furthermore, the increase in Cav1.2 proteins caused by UBR3/6 reduction was not counteracted by a protein biosynthesis inhibitor, cycloheximide, suggesting a degradative regulation of UBR3/6 on Cav1.2 channels. Our results indicate that UBR3/6 modulates cardiac CICR via targeting Cav1.2 protein degradation.
Collapse
Affiliation(s)
- Xiu-E Ma
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine , Shanghai, China
| | - Bei Liu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University , Shanghai, China
| | - Chun-Xia Zhao
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University , Shanghai, China
| |
Collapse
|
17
|
Lory P, Nicole S, Monteil A. Neuronal Cav3 channelopathies: recent progress and perspectives. Pflugers Arch 2020; 472:831-844. [PMID: 32638069 PMCID: PMC7351805 DOI: 10.1007/s00424-020-02429-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
T-type, low-voltage activated, calcium channels, now designated Cav3 channels, are involved in a wide variety of physiological functions, especially in nervous systems. Their unique electrophysiological properties allow them to finely regulate neuronal excitability and to contribute to sensory processing, sleep, and hormone and neurotransmitter release. In the last two decades, genetic studies, including exploration of knock-out mouse models, have greatly contributed to elucidate the role of Cav3 channels in normal physiology, their regulation, and their implication in diseases. Mutations in genes encoding Cav3 channels (CACNA1G, CACNA1H, and CACNA1I) have been linked to a variety of neurodevelopmental, neurological, and psychiatric diseases designated here as neuronal Cav3 channelopathies. In this review, we describe and discuss the clinical findings and supporting in vitro and in vivo studies of the mutant channels, with a focus on de novo, gain-of-function missense mutations recently discovered in CACNA1G and CACNA1H. Overall, the studies of the Cav3 channelopathies help deciphering the pathogenic mechanisms of corresponding diseases and better delineate the properties and physiological roles Cav3 channels.
Collapse
Affiliation(s)
- Philippe Lory
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France. .,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France.
| | - Sophie Nicole
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France
| | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France
| |
Collapse
|
18
|
Marcantoni A, Calorio C, Hidisoglu E, Chiantia G, Carbone E. Cav1.2 channelopathies causing autism: new hallmarks on Timothy syndrome. Pflugers Arch 2020; 472:775-789. [PMID: 32621084 DOI: 10.1007/s00424-020-02430-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Cav1.2 L-type calcium channels play key roles in long-term synaptic plasticity, sensory transduction, muscle contraction, and hormone release. De novo mutations in the gene encoding Cav1.2 (CACNA1C) causes two forms of Timothy syndrome (TS1, TS2), characterized by a multisystem disorder inclusive of cardiac arrhythmias, long QT, autism, and adrenal gland dysfunction. In both TS1 and TS2, the missense mutation G406R is on the alternatively spliced exon 8 and 8A coding for the IS6-helix of Cav1.2 and is responsible for the penetrant form of autism in most TS individuals. The mutation causes specific gain-of-function changes to Cav1.2 channel gating: a "leftward shift" of voltage-dependent activation, reduced voltage-dependent inactivation, and a "leftward shift" of steady-state inactivation. How this occurs and how Cav1.2 gating changes alter neuronal firing and synaptic plasticity is still largely unexplained. Trying to better understanding the molecular basis of Cav1.2 gating dysfunctions leading to autism, here, we will present and discuss the properties of recently reported typical and atypical TS phenotypes and the effective gating changes exhibited by missense mutations associated with long QTs without extracardiac symptoms, unrelated to TS. We will also discuss new emerging views achieved from using iPSCs-derived neurons and the newly available autistic TS2-neo mouse model, both appearing promising for understanding neuronal mistuning in autistic TS patients. We will also analyze and describe recent proposals of molecular pathways that might explain mistuned Ca2+-mediated and Ca2+-independent excitation-transcription signals to the nucleus. Briefly, we will also discuss possible pharmacological approaches to treat autism associated with L-type channelopathies.
Collapse
Affiliation(s)
- Andrea Marcantoni
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Chiara Calorio
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Enis Hidisoglu
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Giuseppe Chiantia
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy
| | - Emilio Carbone
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, N.I.S. Centre, Corso Raffaello 30, 10125, Torino, Italy.
| |
Collapse
|
19
|
Ortner NJ, Kaserer T, Copeland JN, Striessnig J. De novo CACNA1D Ca 2+ channelopathies: clinical phenotypes and molecular mechanism. Pflugers Arch 2020; 472:755-773. [PMID: 32583268 PMCID: PMC7351864 DOI: 10.1007/s00424-020-02418-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
The identification of rare disease-causing variants in humans by large-scale next-generation sequencing (NGS) studies has also provided us with new insights into the pathophysiological role of de novo missense variants in the CACNA1D gene that encodes the pore-forming α1-subunit of voltage-gated Cav1.3 L-type Ca2+ channels. These CACNA1D variants have been identified somatically in aldosterone-producing adenomas as well as germline in patients with neurodevelopmental and in some cases endocrine symptoms. In vitro studies in heterologous expression systems have revealed typical gating changes that indicate enhanced Ca2+ influx through Cav1.3 channels as the underlying disease-causing mechanism. Here we summarize the clinical findings of 12 well-characterized individuals with a total of 9 high-risk pathogenic CACNA1D variants. Moreover, we propose how information from somatic mutations in aldosterone-producing adenomas could be used to predict the potential pathogenicity of novel germline variants. Since these pathogenic de novo variants can cause a channel-gain-of function, we also discuss the use of L-type Ca2+ channel blockers as a potential therapeutic option.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Teresa Kaserer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - J Nathan Copeland
- Duke Center for Autism and Brain Development, Duke Child and Family Mental Health and Developmental Neuroscience, Durham, USA
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
20
|
Cummings BB, Karczewski KJ, Kosmicki JA, Seaby EG, Watts NA, Singer-Berk M, Mudge JM, Karjalainen J, Satterstrom FK, O'Donnell-Luria AH, Poterba T, Seed C, Solomonson M, Alföldi J, Daly MJ, MacArthur DG. Transcript expression-aware annotation improves rare variant interpretation. Nature 2020; 581:452-458. [PMID: 32461655 PMCID: PMC7334198 DOI: 10.1038/s41586-020-2329-2] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/23/2020] [Indexed: 01/09/2023]
Abstract
The acceleration of DNA sequencing in samples from patients and population studies has resulted in extensive catalogues of human genetic variation, but the interpretation of rare genetic variants remains problematic. A notable example of this challenge is the existence of disruptive variants in dosage-sensitive disease genes, even in apparently healthy individuals. Here, by manual curation of putative loss-of-function (pLoF) variants in haploinsufficient disease genes in the Genome Aggregation Database (gnomAD)1, we show that one explanation for this paradox involves alternative splicing of mRNA, which allows exons of a gene to be expressed at varying levels across different cell types. Currently, no existing annotation tool systematically incorporates information about exon expression into the interpretation of variants. We develop a transcript-level annotation metric known as the 'proportion expressed across transcripts', which quantifies isoform expression for variants. We calculate this metric using 11,706 tissue samples from the Genotype Tissue Expression (GTEx) project2 and show that it can differentiate between weakly and highly evolutionarily conserved exons, a proxy for functional importance. We demonstrate that expression-based annotation selectively filters 22.8% of falsely annotated pLoF variants found in haploinsufficient disease genes in gnomAD, while removing less than 4% of high-confidence pathogenic variants in the same genes. Finally, we apply our expression filter to the analysis of de novo variants in patients with autism spectrum disorder and intellectual disability or developmental disorders to show that pLoF variants in weakly expressed regions have similar effect sizes to those of synonymous variants, whereas pLoF variants in highly expressed exons are most strongly enriched among cases. Our annotation is fast, flexible and generalizable, making it possible for any variant file to be annotated with any isoform expression dataset, and will be valuable for the genetic diagnosis of rare diseases, the analysis of rare variant burden in complex disorders, and the curation and prioritization of variants in recall-by-genotype studies.
Collapse
Affiliation(s)
- Beryl B Cummings
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Konrad J Karczewski
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Jack A Kosmicki
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA, USA
| | - Eleanor G Seaby
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Genomic Informatics Group, University Hospital Southampton, Southampton, UK
| | - Nicholas A Watts
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Moriel Singer-Berk
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan M Mudge
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Juha Karjalainen
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - F Kyle Satterstrom
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne H O'Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy Poterba
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cotton Seed
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew Solomonson
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Alföldi
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Mark J Daly
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel G MacArthur
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Centre for Population Genomics, Garvan Institute of Medical Research, and UNSW Sydney, Syndney, Australia.
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Australia.
| |
Collapse
|
21
|
Nayeri T, Sarvi S, Moosazadeh M, Hosseininejad Z, Amouei A, Daryani A. Toxoplasma gondii infection and risk of attention-deficit hyperactivity disorder: a systematic review and meta-analysis. Pathog Glob Health 2020; 114:117-126. [PMID: 32186992 DOI: 10.1080/20477724.2020.1738153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Toxoplasma gondii (T. gondii), as an opportunistic neurotropic parasite of the Apicomplexa family, was firstly described in 1908. As attention-deficit hyperactivity disorder (ADHD) is one of the most common neuropsychiatric disorders in children and adolescents and often persists into adulthood, the purpose of this systematic review and meta-analysis was to investigate the relationship between T. gondii infection and ADHD.The data were systematically collected from seven electronic databases up to May 1st 2019 with no language restriction. This study was registered at the International Prospective Register of Systematic Reviews (PROSPERO; code: CRD42020149353). Odds ratios (ORs) and 95% confidence intervals (CI) were estimated using a random effects model. Seven studies involving five cross-sectional and two case-control studies were included in this meta-analysis.Results indicated that there was a statistically non-significant association between exposure to T. gondii infection and increased risk of ADHD based on the detection of immunoglobulin G (IgG) antibody (2.02 [95% CI: 0.97-4.20]; I2=58.7%). However, obtained results of Egger's tests for anti-T. gondii IgG antibody showed publication bias (P=0.014).Sensitivity analysis revealed stable results for the association between anti-T. gondii IgG antibody with ADHD.Given the small number of studies in this field and the obtained results, it cannot be conclusively stated that T. gondii is a risk factor for ADHD.It is important to have reliable information about the relationship between T. gondii and ADHD around the world; as it may lead to better insight to elucidate the possible association of toxoplasmosis and the pathogenesis of ADHD.
Collapse
Affiliation(s)
- Tooran Nayeri
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahmood Moosazadeh
- Health Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Hosseininejad
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Afsaneh Amouei
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
22
|
Bhandari R, Paliwal JK, Kuhad A. Neuropsychopathology of Autism Spectrum Disorder: Complex Interplay of Genetic, Epigenetic, and Environmental Factors. ADVANCES IN NEUROBIOLOGY 2020; 24:97-141. [PMID: 32006358 DOI: 10.1007/978-3-030-30402-7_4] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Autism spectrum disorder (ASD) is a complex heterogeneous consortium of pervasive development disorders (PDD) which ranges from atypical autism, autism, and Asperger syndrome affecting brain in the developmental stage. This debilitating neurodevelopmental disorder results in both core as well as associated symptoms. Core symptoms observed in autistic patients are lack of social interaction, pervasive, stereotyped, and restricted behavior while the associated symptoms include irritability, anxiety, aggression, and several comorbid disorders.ASD is a polygenic disorder and is multifactorial in origin. Copy number variations (CNVs) of several genes that regulate the synaptogenesis and signaling pathways are one of the major factors responsible for the pathogenesis of autism. The complex integration of various CNVs cause mutations in the genes which code for molecules involved in cell adhesion, voltage-gated ion-channels, scaffolding proteins as well as signaling pathways (PTEN and mTOR pathways). These mutated genes are responsible for affecting synaptic transmission by causing plasticity dysfunction responsible, in turn, for the expression of ASD.Epigenetic modifications affecting DNA transcription and various pre-natal and post-natal exposure to a variety of environmental factors are also precipitating factors for the occurrence of ASD. All of these together cause dysregulation of glutamatergic signaling as well as imbalance in excitatory: inhibitory pathways resulting in glial cell activation and release of inflammatory mediators responsible for the aberrant social behavior which is observed in autistic patients.In this chapter we review and provide insight into the intricate integration of various genetic, epigenetic, and environmental factors which play a major role in the pathogenesis of this disorder and the mechanistic approach behind this integration.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Jyoti K Paliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India.
| |
Collapse
|
23
|
Sutterland AL, Blom MT, Ladee K, Lubbers JJM, Cohen D, de Haan L, Tan HL. Increased prevalence of ECG suspicious for Brugada Syndrome in recent onset schizophrenia spectrum disorders. Schizophr Res 2019; 210:59-65. [PMID: 31248748 DOI: 10.1016/j.schres.2019.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/29/2019] [Accepted: 06/17/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Schizophrenia is associated with an increased risk of sudden cardiac death, traditionally attributed to prolonged QTc interval and increased prevalence of cardiovascular risk factors. However, defective ion channels implicated in both schizophrenia and Brugada Syndrome (BrS) may be associated with an increased risk of cardiac arrhythmias. Moreover, these cardiac arrhythmias can be provoked by various drugs, including psychotropic drugs. OBJECTIVE To assess the prevalence of the occurrence of ECG suspicious for BrS (suspect BrS-ECG) and the prevalence of BrS in patients with recent onset schizophrenia spectrum disorders (SSD). METHODS In this case-control study, ECGs of 388 patients with recent onset SSD admitted between 2006 and 2015 and 844 healthy controls were made. All persons who had a suspect BrS-ECG were offered an ajmaline provocation test to diagnose or exclude BrS. Data on possible confounders were ascertained. Patients with and without suspect BrS-ECG were compared regarding clinical and ECG variables. RESULTS Suspect BrS-ECG was found in 33 patients (8.5%) and 13 healthy controls (1.5%), with an adjusted Odds Ratio of 3.5 (p < 0.0001). This finding was not explained by potential confounders such as gender, age, ethnicity, cannabis use, cardiovascular risk factors, medication use or serum electrolytes. BrS was confirmed in three patients and one control. CONCLUSION A considerable subset of patients with recent onset SSD have suspect BrS-ECG, extending earlier findings in patients with chronic schizophrenia. Screening for BrS in schizophrenia could be relevant both to prevent sudden cardiac death and to identify a subgroup of patients with possible ion-channel dysfunctioning.
Collapse
Affiliation(s)
- Arjen L Sutterland
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Early Psychosis Section, Amsterdam, the Netherlands.
| | - Marieke T Blom
- Amsterdam UMC, University of Amsterdam, Department of Cardiology, Heart Center, Amsterdam, the Netherlands
| | - Katinka Ladee
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Early Psychosis Section, Amsterdam, the Netherlands
| | - Jorieke J M Lubbers
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Early Psychosis Section, Amsterdam, the Netherlands
| | - Dan Cohen
- Mental Health Service North-Holland North, Department of Community Mental Health, Heerhugowaard, the Netherlands
| | - Lieuwe de Haan
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Early Psychosis Section, Amsterdam, the Netherlands
| | - Hanno L Tan
- Amsterdam UMC, University of Amsterdam, Department of Cardiology, Heart Center, Amsterdam, the Netherlands
| |
Collapse
|
24
|
Nanou E, Catterall WA. Calcium Channels, Synaptic Plasticity, and Neuropsychiatric Disease. Neuron 2019; 98:466-481. [PMID: 29723500 DOI: 10.1016/j.neuron.2018.03.017] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/06/2018] [Accepted: 03/09/2018] [Indexed: 12/14/2022]
Abstract
Voltage-gated calcium channels couple depolarization of the cell-surface membrane to entry of calcium, which triggers secretion, contraction, neurotransmission, gene expression, and other physiological responses. They are encoded by ten genes, which generate three voltage-gated calcium channel subfamilies: CaV1; CaV2; and CaV3. At synapses, CaV2 channels form large signaling complexes in the presynaptic nerve terminal, which are responsible for the calcium entry that triggers neurotransmitter release and short-term presynaptic plasticity. CaV1 channels form signaling complexes in postsynaptic dendrites and dendritic spines, where their calcium entry induces long-term potentiation. These calcium channels are the targets of mutations and polymorphisms that alter their function and/or regulation and cause neuropsychiatric diseases, including migraine headache, cerebellar ataxia, autism, schizophrenia, bipolar disorder, and depression. This article reviews the molecular properties of calcium channels, considers their multiple roles in synaptic plasticity, and discusses their potential involvement in this wide range of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Evanthia Nanou
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA
| | - William A Catterall
- Department of Pharmacology, University of Washington, Seattle, WA 98195-7280, USA.
| |
Collapse
|
25
|
Du X, Wei C, Hejazi Pastor DP, Rao ER, Li Y, Grasselli G, Godfrey J, Palmenberg AC, Andrade J, Hansel C, Gomez CM. α1ACT Is Essential for Survival and Early Cerebellar Programming in a Critical Neonatal Window. Neuron 2019; 102:770-785.e7. [PMID: 30922876 DOI: 10.1016/j.neuron.2019.02.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 12/17/2018] [Accepted: 02/20/2019] [Indexed: 10/27/2022]
Abstract
Postnatal cerebellar development is a precisely regulated process involving well-orchestrated expression of neural genes. Neurological phenotypes associated with CACNA1A gene defects have been increasingly recognized, yet the molecular principles underlying this association remain elusive. By characterizing a dose-dependent CACNA1A gene deficiency mouse model, we discovered that α1ACT, as a transcription factor and secondary protein of CACNA1A mRNA, drives dynamic gene expression networks within cerebellar Purkinje cells and is indispensable for neonatal survival. Perinatal loss of α1ACT leads to motor dysfunction through disruption of neurogenesis and synaptic regulatory networks. However, its elimination in adulthood has minimal effect on the cerebellum. These findings shed light on the critical role of α1ACT in facilitating neuronal development in both mice and humans and support a rationale for gene therapies for calcium-channel-associated cerebellar disorders. Finally, we show that bicistronic expression may be common to the voltage-gated calcium channel (VGCC) gene family and may help explain complex genetic syndromes.
Collapse
Affiliation(s)
- Xiaofei Du
- Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Cenfu Wei
- Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | | | - Eshaan R Rao
- Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Yan Li
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA
| | - Giorgio Grasselli
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA; Center for Synaptic Neuroscience and Technology, Italian Institute of Technology (IIT), L.go R. Benzi 10, 16132 Genova, Italy
| | - Jack Godfrey
- Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Ann C Palmenberg
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jorge Andrade
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA; Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | - Christian Hansel
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
26
|
Clinical Outcomes and Modes of Death in Timothy Syndrome. JACC Clin Electrophysiol 2018; 4:459-466. [DOI: 10.1016/j.jacep.2017.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 11/19/2022]
|
27
|
STAC proteins associate to the IQ domain of Ca V1.2 and inhibit calcium-dependent inactivation. Proc Natl Acad Sci U S A 2018; 115:1376-1381. [PMID: 29363593 PMCID: PMC5819422 DOI: 10.1073/pnas.1715997115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Association of the adaptor protein STAC3 to the calcium channel was recently identified as essential for skeletal muscle contraction. While STAC3 is expressed exclusively in skeletal muscle, the other two isoforms, STAC1 and STAC2, are found in the brain, but their function has not yet been investigated. Recently we identified the C1 domain of STAC as crucial for its association with calcium channels. However, the corresponding binding domain remained unknown. Here, we demonstrate that STAC proteins associate with the C terminus of CaV1.2 at the IQ domain, and that this interaction results in the inhibition of calcium-dependent inactivation. Taken together, our data identify a functionally important STAC interaction domain and suggest that STAC proteins modulate calcium entry through CaV1 channels. The adaptor proteins STAC1, STAC2, and STAC3 represent a newly identified family of regulators of voltage-gated calcium channel (CaV) trafficking and function. The skeletal muscle isoform STAC3 is essential for excitation–contraction coupling and its mutation causes severe muscle disease. Recently, two distinct molecular domains in STAC3 were identified, necessary for its functional interaction with CaV1.1: the C1 domain, which recruits STAC proteins to the calcium channel complex in skeletal muscle triads, and the SH3-1 domain, involved in excitation–contraction coupling. These interaction sites are conserved in the three STAC proteins. However, the molecular domain in CaV1 channels interacting with the STAC C1 domain and the possible role of this interaction in neuronal CaV1 channels remained unknown. Using CaV1.2/2.1 chimeras expressed in dysgenic (CaV1.1−/−) myotubes, we identified the amino acids 1,641–1,668 in the C terminus of CaV1.2 as necessary for association of STAC proteins. This sequence contains the IQ domain and alanine mutagenesis revealed that the amino acids important for STAC association overlap with those making contacts with the C-lobe of calcium-calmodulin (Ca/CaM) and mediating calcium-dependent inactivation of CaV1.2. Indeed, patch-clamp analysis demonstrated that coexpression of either one of the three STAC proteins with CaV1.2 opposed calcium-dependent inactivation, although to different degrees, and that substitution of the CaV1.2 IQ domain with that of CaV2.1, which does not interact with STAC, abolished this effect. These results suggest that STAC proteins associate with the CaV1.2 C terminus at the IQ domain and thus inhibit calcium-dependent feedback regulation of CaV1.2 currents.
Collapse
|
28
|
González-Ramírez R, Felix R. Transcriptional regulation of voltage-gated Ca 2+ channels. Acta Physiol (Oxf) 2018; 222. [PMID: 28371478 DOI: 10.1111/apha.12883] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/14/2017] [Accepted: 03/21/2017] [Indexed: 12/30/2022]
Abstract
The transcriptional regulation of voltage-gated Ca2+ (CaV ) channels is an emerging research area that promises to improve our understanding of how many relevant physiological events are shaped in the central nervous system, the skeletal muscle and other tissues. Interestingly, a picture of how transcription of CaV channel subunit genes is controlled is evolving with the identification of the promoter regions required for tissue-specific expression and the identification of transcription factors that control their expression. These promoters share several characteristics that include multiple transcriptional start sites, lack of a TATA box and the presence of elements conferring tissue-selective expression. Likewise, changes in CaV channel expression occur throughout development, following ischaemia, seizures or chronic drug administration. This review focuses on insights achieved regarding the control of CaV channel gene expression. To further understand the complexities of expression and to increase the possibilities of detecting CaV channel alterations causing human disease, a deeper knowledge on the structure of the 5' upstream regions of the genes encoding these remarkable proteins will be necessary.
Collapse
Affiliation(s)
- R. González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad; Hospital General ‘Dr. Manuel Gea González’; Secretaría de Salud; Ciudad de México México
| | - R. Felix
- Departmento de Biología Celular; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN); Ciudad de México México
| |
Collapse
|
29
|
Zhang Q, Chen J, Qin Y, Wang J, Zhou L. Mutations in voltage-gated L-type calcium channel: implications in cardiac arrhythmia. Channels (Austin) 2018; 12:201-218. [PMID: 30027834 PMCID: PMC6104696 DOI: 10.1080/19336950.2018.1499368] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/08/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023] Open
Abstract
The voltage-gated L-type calcium channel (LTCC) is essential for multiple cellular processes. In the heart, calcium influx through LTCC plays an important role in cardiac electrical excitation. Mutations in LTCC genes, including CACNA1C, CACNA1D, CACNB2 and CACNA2D, will induce the dysfunctions of calcium channels, which result in the abnormal excitations of cardiomyocytes, and finally lead to cardiac arrhythmias. Nevertheless, the newly found mutations in LTCC and their functions are continuously being elucidated. This review summarizes recent findings on the mutations of LTCC, which are associated with long QT syndromes, Timothy syndromes, Brugada syndromes, short QT syndromes, and some other cardiac arrhythmias. Indeed, we describe the gain/loss-of-functions of these mutations in LTCC, which can give an explanation for the phenotypes of cardiac arrhythmias. Moreover, we present several challenges in the field at present, and propose some diagnostic or therapeutic approaches to these mutation-associated cardiac diseases in the future.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, the Second Affiliated Hospital of Nantong University, Nantong First Hospital, Nantong, Jiangsu, China
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junjie Chen
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yao Qin
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
30
|
Völkening B, Schönig K, Kronenberg G, Bartsch D, Weber T. Type-1 astrocyte-like stem cells harboring Cacna1d gene deletion exhibit reduced proliferation and decreased neuronal fate choice. Hippocampus 2017; 28:97-107. [PMID: 29116659 DOI: 10.1002/hipo.22811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/26/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022]
Abstract
In the central nervous system, CaV 1.2 and CaV 1. 3 constitute the main L-type voltage-gated calcium channels (LTCCs) coupling membrane depolarization to gene transcription. We have previously demonstrated that inducible disruption of Cav1.2 in type-1 astrocyte-like stem cells of the adult dentate gyrus (DG) impairs hippocampal neurogenesis in a cell-autonomous fashion. To address the role of Cav1.3 channels (encoded by the Cacna1d gene), we here generated TgGLAST-CreERT2 /Cacna1dfl/fl /RCE:loxP mice which facilitate inducible deletion of Cacna1d in tandem with induction of EGFP expression in type-1 cells, allowing tracking of recombined cells and their descendants. Neurosphere cultures derived from fluorescence-activated cell sorting sorted Cacna1d-deficient (Cacna1d-/- /EGFP) hippocampal neural precursor cells (NPCs) exhibited a significant decrease in proliferative activity. Further, under differentiation conditions, Cacna1d deficiency conferred an increase in astrogenesis at the expense of neurogenesis. In like manner, type-1 cells lacking Cacna1d showed reduced proliferation in the dentate gyrus (DG) in vivo. Moreover, Cacna1d deficiency resulted in a significant decrease in the number of newly born cells adopting a neuronal fate. Finally, massive excitation induced by repeated electroconvulsive seizures rescued the proliferation defect of Cacna1d-/- /EGFP type-1 cells. Together, the effects of Cacna1d gene deletion closely recapitulate our earlier findings on the role of Cav1.2 channels expressed by type-1 cells. Similar to Cav1.2 channels, Cav1.3 channels on type-1 cells boost type-1 cell proliferation and promote subsequent neuronal fate choice.
Collapse
Affiliation(s)
- Bianca Völkening
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, Mannheim 68159, Germany
| | - Kai Schönig
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, Mannheim 68159, Germany
| | - Golo Kronenberg
- Department of Psychiatry and Psychotherapy, University of Rostock, Gehlsheimer Straße 20, Rostock 18147, Germany
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, Mannheim 68159, Germany
| | - Tillmann Weber
- Department of Molecular Biology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, Mannheim 68159, Germany.,Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, Mannheim 68159, Germany
| |
Collapse
|
31
|
Berridge MJ. Vitamin D deficiency: infertility and neurodevelopmental diseases (attention deficit hyperactivity disorder, autism, and schizophrenia). Am J Physiol Cell Physiol 2017; 314:C135-C151. [PMID: 29070492 DOI: 10.1152/ajpcell.00188.2017] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The process of development depends on a number of signaling systems that regulates the progressive sequence of developmental events. Infertility and neurodevelopmental diseases, such as attention deficit hyperactivity disorder, autism spectrum disorders, and schizophrenia, are caused by specific alterations in these signaling processes. Calcium signaling plays a prominent role throughout development beginning at fertilization and continuing through early development, implantation, and organ differentiation such as heart and brain development. Vitamin D plays a major role in regulating these signaling processes that control development. There is an increase in infertility and an onset of neurodevelopmental diseases when vitamin D is deficient. The way in which vitamin D deficiency acts to alter development is a major feature of this review. One of the primary functions of vitamin D is to maintain the phenotypic stability of both the Ca2+ and redox signaling pathways that play such a key role throughout development.
Collapse
Affiliation(s)
- Michael J Berridge
- Laboratory of Molecular Signalling, The Babraham Institute , Cambridge , United Kingdom
| |
Collapse
|
32
|
Sepp R, Hategan L, Bácsi A, Cseklye J, Környei L, Borbás J, Széll M, Forster T, Nagy I, Hegedűs Z. Timothy syndrome 1 genotype without syndactyly and major extracardiac manifestations. Am J Med Genet A 2017; 173:784-789. [PMID: 28211989 DOI: 10.1002/ajmg.a.38084] [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: 08/29/2016] [Accepted: 11/21/2016] [Indexed: 11/09/2022]
Abstract
Timothy syndrome 1 (TS1) is a rare genetic disorder characterized by multisystem abnormalities including QT prolongation, congenital heart defects, facial dysmorphism, episodic hypoglycemia, and neurological symptoms. A morphological hallmark of TS1 is syndactyly, present in all cases. TS1 is caused by the canonical p.Gly406Arg mutation in the alternatively spliced exon 8A in the CACNA1C gene, encoding for the main cardiac L-type calcium channel. A variant case of TS1 is reported. The proband had intermittent fetal bradycardia with heart rate of 72 bpm. On the first day of life bradycardia due to 2:1 atrioventricular (AV) block and marked QTc prolongation of 600 ms was noted. On medical therapy with propranolol and mexiletine 1:1 AV conduction returned with QTc prolongation of 470-580 ms. The patient lacked other extracardiac manifestations, most importantly syndactyly, neurological complications or autism. On genetic analysis, the canonical TS1 causing mutation, p.Gly406Arg in exon 8A of the CACNA1C gene was detected. The CACNA1C p.Gly406Arg variant was not present in the parents, but was detected in different DNA samples of the index patient. Our case highlight further phenotypic variability in TS. Most importantly, it underlines that the lack of syndactyly does not exclude the presence of a TS1 genotype. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Róbert Sepp
- Second Department of Internal Medicine and Cardiology Center, University of Szeged, Szeged, Hungary
| | - Lidia Hategan
- Second Department of Internal Medicine and Cardiology Center, University of Szeged, Szeged, Hungary
| | - Attila Bácsi
- Fejér County "Szent György" University Teaching Hospital, Székesfehérvár, Hungary
| | | | - László Környei
- "Gottsegen György" National Institute of Cardiology, Budapest, Hungary
| | - János Borbás
- Second Department of Internal Medicine and Cardiology Center, University of Szeged, Szeged, Hungary
| | - Márta Széll
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - Tamás Forster
- Second Department of Internal Medicine and Cardiology Center, University of Szeged, Szeged, Hungary
| | - István Nagy
- Seqomics Biotechnology Ltd., Mórahalom, Hungary.,Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Zoltán Hegedűs
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Department of Biochemistry and Medical Chemistry, University of Pécs, Pécs, Hungary
| |
Collapse
|
33
|
Rai V, Agrawal DK. Role of risk stratification and genetics in sudden cardiac death. Can J Physiol Pharmacol 2016; 95:225-238. [PMID: 27875062 DOI: 10.1139/cjpp-2016-0457] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sudden cardiac death (SCD) is a major public health issue due to its increasing incidence in the general population and the difficulty in identifying high-risk individuals. Nearly 300 000 - 350 000 patients in the United States and 4-5 million patients in the world die annually from SCD. Coronary artery disease and advanced heart failure are the main etiology for SCD. Ischemia of any cause precipitates lethal arrhythmias, and ventricular tachycardia and ventricular fibrillation are the most common lethal arrhythmias precipitating SCD. Pulseless electrical activity, bradyarrhythmia, and electromechanical dissociation also result in SCD. Most SCDs occur outside of the hospital setting, so it is difficult to estimate the public burden, which results in overestimating the incidence of SCD. The insufficiency and limited predictive value of various indicators and criteria for SCD result in the increasing incidence. As a result, there is a need to develop better risk stratification criteria and find modifiable variables to decrease the incidence. Primary and secondary prevention and treatment of SCD need further research. This critical review is focused on the etiology, risk factors, prognostic factors, and importance of risk stratification of SCD.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA.,Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| |
Collapse
|
34
|
Rao S, Yao Y, Zheng C, Ryan J, Mao C, Zhang F, Meyre D, Xu Q. Common variants in CACNA1C and MDD susceptibility: A comprehensive meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2016; 171:896-903. [PMID: 27260792 DOI: 10.1002/ajmg.b.32466] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/20/2016] [Indexed: 01/11/2023]
Abstract
Major depressive disorder (MDD) is one of the most common psychiatric disorders with a relatively high heritability (35-40%). Though rs1006737 in the CACNA1C gene showed significant association with MDD in a British large-scale candidate association study, most of the replication analyses with relatively small sample size reported negative association. Moreover, this locus has never been identified in previous genome-wide association studies (GWAS) for MDD. Here, we conducted a comprehensive meta-analysis of the association between CACNA1C variants and MDD risk by combining all published data. Genetic data from one European GWAS and five individual follow-up studies, which include up to 12,629 patients of MDD and 28,653 controls, that is, the largest sample size on CACNA1C to date, were collected. Rs1006737 showed significant association with MDD in the fixed-effect model (Z = 2.56, P = 0.011, OR = 1.08, 95%CI = 1.04-1.12) and the association remained after reanalyzing the data according to ethnicity. We additionally analyzed other 25 SNPs, genotyped in only one replication study, across the CACNA1C locus, and found that two SNPs, rs4765905 (P = 0.041, OR = 1.05, 95%CI 1.00-1.09) and rs4765937 (P = 0.025, OR = 1.05, 95%CI 1.01-1.09) showed nominal association with MDD, while rs2239073 (P = 0.002, OR = 1.07, 95%CI 1.02-1.11) exhibited significant association with MDD, which survived from multiple corrections. Our study provides support for positive association between CACNA1C and MDD; however, the current data suggest the necessity of replication analyses in a larger-scale sample. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shuquan Rao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yao Yao
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Zheng
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Joanne Ryan
- Disease Epigenetics Group, Murdoch Children Research Institute and Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Inserm, U1061, Univ Montpellier, Montpellier, France
| | - Canquan Mao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Fuquan Zhang
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Qi Xu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Prieto ML, Ryu E, Jenkins GD, Batzler A, Nassan MM, Cuellar-Barboza AB, Pathak J, McElroy SL, Frye MA, Biernacka JM. Leveraging electronic health records to study pleiotropic effects on bipolar disorder and medical comorbidities. Transl Psychiatry 2016; 6:e870. [PMID: 27529678 PMCID: PMC5022084 DOI: 10.1038/tp.2016.138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 05/13/2016] [Accepted: 06/15/2016] [Indexed: 01/27/2023] Open
Abstract
Patients with bipolar disorder (BD) have a high prevalence of comorbid medical illness. However, the mechanisms underlying these comorbidities with BD are not well known. Certain genetic variants may have pleiotropic effects, increasing the risk of BD and other medical illnesses simultaneously. In this study, we evaluated the association of BD-susceptibility genetic variants with various medical conditions that tend to co-exist with BD, using electronic health records (EHR) data linked to genome-wide single-nucleotide polymorphism (SNP) data. Data from 7316 Caucasian subjects were used to test the association of 19 EHR-derived phenotypes with 34 SNPs that were previously reported to be associated with BD. After Bonferroni multiple testing correction, P<7.7 × 10(-5) was considered statistically significant. The top association findings suggested that the BD risk alleles at SNP rs4765913 in CACNA1C gene and rs7042161 in SVEP1 may be associated with increased risk of 'cardiac dysrhythmias' (odds ratio (OR)=1.1, P=3.4 × 10(-3)) and 'essential hypertension' (OR=1.1, P=3.5 × 10(-3)), respectively. Although these associations are not statistically significant after multiple testing correction, both genes have been previously implicated with cardiovascular phenotypes. Moreover, we present additional evidence supporting these associations, particularly the association of the SVEP1 SNP with hypertension. This study shows the potential for EHR-based analyses of large cohorts to discover pleiotropic effects contributing to complex psychiatric traits and commonly co-occurring medical conditions.
Collapse
Affiliation(s)
- M L Prieto
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Universidad de los Andes, Facultad de Medicina, Departamento de Psiquiatría, Santiago, Chile
| | - E Ryu
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - G D Jenkins
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A Batzler
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - M M Nassan
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A B Cuellar-Barboza
- Department of Psychiatry, Universidad Autónoma de Nuevo León, Nuevo León, Mexico
| | - J Pathak
- Division of Health Informatics, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - S L McElroy
- Lindner Center of HOPE, Mason, OH, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - M A Frye
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - J M Biernacka
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
36
|
Yang ZL, Sun GL. [Research advances in candidate genes for autism spectrum disorder]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:282-287. [PMID: 26975830 PMCID: PMC7390002 DOI: 10.7499/j.issn.1008-8830.2016.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/15/2015] [Indexed: 06/05/2023]
Abstract
Autism spectrum disorder (ASD) is a kind of neurodevelopmental multigenic disorder. More than one hundred of candidate genes for ASD have been reported. The candidate gene research for ASD involves in chromosome loci and screening of candidate genes and epigenetic abnormalities for candidate genes. The reported genes encode neural adhesion molecules, ion channels, scaffold proteins, protein kinases, receptor protein and carrier protein, signaling modulate molecules and circadian relevant proteins. The research of mutation screening and expression regulation of candidate genes can help to elucidate genetic mechanisms for ASD, and may provide new approaches for the diagnosis and treatment of this disorder. This article reviews the research advance in candidate genes for ASD.
Collapse
Affiliation(s)
- Zhi-Liang Yang
- Department of Pediatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | | |
Collapse
|
37
|
Dick IE, Joshi-Mukherjee R, Yang W, Yue DT. Arrhythmogenesis in Timothy Syndrome is associated with defects in Ca(2+)-dependent inactivation. Nat Commun 2016; 7:10370. [PMID: 26822303 PMCID: PMC4740114 DOI: 10.1038/ncomms10370] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/03/2015] [Indexed: 12/18/2022] Open
Abstract
Timothy Syndrome (TS) is a multisystem disorder, prominently featuring cardiac action potential prolongation with paroxysms of life-threatening arrhythmias. The underlying defect is a single de novo missense mutation in CaV1.2 channels, either G406R or G402S. Notably, these mutations are often viewed as equivalent, as they produce comparable defects in voltage-dependent inactivation and cause similar manifestations in patients. Yet, their effects on calcium-dependent inactivation (CDI) have remained uncertain. Here, we find a significant defect in CDI in TS channels, and uncover a remarkable divergence in the underlying mechanism for G406R versus G402S variants. Moreover, expression of these TS channels in cultured adult guinea pig myocytes, combined with a quantitative ventricular myocyte model, reveals a threshold behaviour in the induction of arrhythmias due to TS channel expression, suggesting an important therapeutic principle: a small shift in the complement of mutant versus wild-type channels may confer significant clinical improvement. Timothy Syndrome (TS) is a multisystem disorder caused by two mutations leading to dysfunction of the CaV1.2 channel. Here, Dick et al. uncover a major and mechanistically divergent effect of both mutations on Ca2+/calmodulin-dependent inactivation of CaV1.2 channels, suggesting genetic variant-tailored therapy for TS treatment.
Collapse
Affiliation(s)
- 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, Maryland 21205, USA
| | - Rosy Joshi-Mukherjee
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, Maryland 21205, USA
| | - Wanjun Yang
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, Maryland 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, Maryland 21205, USA
| |
Collapse
|
38
|
Martin S, Lazzarini M, Dullin C, Balakrishnan S, Gomes FV, Ninkovic M, El Hady A, Pardo LA, Stühmer W, Del-Bel E. SK3 Channel Overexpression in Mice Causes Hippocampal Shrinkage Associated with Cognitive Impairments. Mol Neurobiol 2016; 54:1078-1091. [PMID: 26803493 PMCID: PMC5310555 DOI: 10.1007/s12035-015-9680-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/23/2015] [Indexed: 12/11/2022]
Abstract
The dysfunction of the small-conductance calcium-activated K+ channel SK3 has been described as one of the factors responsible for the progress of psychoneurological diseases, but the molecular basis of this is largely unknown. This report reveals through use of immunohistochemistry and computational tomography that long-term increased expression of the SK3 small-conductance calcium-activated potassium channel (SK3-T/T) in mice induces a notable bilateral reduction of the hippocampal area (more than 50 %). Histological analysis showed that SK3-T/T mice have cellular disarrangements and neuron discontinuities in the hippocampal formation CA1 and CA3 neuronal layer. SK3 overexpression resulted in cognitive loss as determined by the object recognition test. Electrophysiological examination of hippocampal slices revealed that SK3 channel overexpression induced deficiency of long-term potentiation in hippocampal microcircuits. In association with these results, there were changes at the mRNA levels of some genes involved in Alzheimer’s disease and/or linked to schizophrenia, epilepsy, and autism. Taken together, these features suggest that augmenting the function of SK3 ion channel in mice may present a unique opportunity to investigate the neural basis of central nervous system dysfunctions associated with schizophrenia, Alzheimer’s disease, or other neuropsychiatric/neurodegenerative disorders in this model system. As a more detailed understanding of the role of the SK3 channel in brain disorders is limited by the lack of specific SK3 antagonists and agonists, the results observed in this study are of significant interest; they suggest a new approach for the development of neuroprotective strategies in neuropsychiatric/neurodegenerative diseases with SK3 representing a potential drug target.
Collapse
Affiliation(s)
- Sabine Martin
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075, Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Marcio Lazzarini
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075, Göttingen, Germany
| | - Christian Dullin
- Department of Diagnostic and Interventional Radiology, Georg-August University Medical Center, 37075, Göttingen, Germany
| | - Saju Balakrishnan
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Department of Neuro- and Sensory Physiology, Georg-August University Medical Center, 37073, Göttingen, Germany
| | - Felipe V Gomes
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, 14040-900, Ribeirão Preto, Brazil
| | - Milena Ninkovic
- Department of Neurosurgery, Georg-August University Medical Center, 37075, Göttingen, Germany
| | - Ahmed El Hady
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075, Göttingen, Germany
- Bernstein Focus for Neurotechnology and Bernstein Center for Computational Neuroscience, Göttingen, Germany
- Theoretical Neurophysics, Department of Non-linear Dynamics, Max Planck Institute for Dynamics and Self-Organization, 37077, Göttingen, Germany
- The Interdisciplinary Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
| | - Walter Stühmer
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075, Göttingen, Germany.
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.
- Bernstein Focus for Neurotechnology and Bernstein Center for Computational Neuroscience, Göttingen, Germany.
| | - Elaine Del-Bel
- Department of Morphology, Physiology and Pathology, CNPQ Research 1B (Biophysics, Biochemistry, Pharmacology and Neuroscience), University of São Paulo Dental School of Ribeirão Preto, Avenida do Café 3400, 14040-904, Ribeirão Preto, Brazil.
| |
Collapse
|
39
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 786] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
40
|
Davenport B, Li Y, Heizer JW, Schmitz C, Perraud AL. Signature Channels of Excitability no More: L-Type Channels in Immune Cells. Front Immunol 2015; 6:375. [PMID: 26257741 PMCID: PMC4512153 DOI: 10.3389/fimmu.2015.00375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/09/2015] [Indexed: 12/16/2022] Open
Abstract
Although the concept of Ca(2+) as a universal messenger is well established, it was assumed that the regulatory mechanisms of Ca(2+)-signaling were divided along the line of electric excitability. Recent advances in molecular biology and genomics have, however, provided evidence that non-excitable cells such as immunocytes also express a wide and diverse pool of ion channels that does not differ as significantly from that of excitable cells as originally assumed. Ion channels and transporters are involved in virtually all aspects of immune response regulation, from cell differentiation and development to activation, and effector functions such as migration, antibody-secretion, phagosomal maturation, or vesicular delivery of bactericidal agents. This comprises TRP channel family members, voltage- and Ca(2+)-gated K(+)- and Na(+)-channels, as well as unexpectedly, components of the CaV1-subfamily of voltage-gated L-type Ca(2+)-channels, originally thought to be signature molecules of excitability. This article provides an overview of recent observations made in the field of CaV1 L-type channel function in the immune context, as well as presents results we obtained studying these channels in B-lymphocytes.
Collapse
Affiliation(s)
- Bennett Davenport
- Department of Biomedical Research, National Jewish Health , Denver, CO , USA ; Department of Immunology and Microbiology, University of Colorado Denver , Denver, CO , USA
| | - Yuan Li
- Department of Biomedical Research, National Jewish Health , Denver, CO , USA ; Department of Immunology and Microbiology, University of Colorado Denver , Denver, CO , USA
| | - Justin W Heizer
- Department of Biomedical Research, National Jewish Health , Denver, CO , USA ; Department of Immunology and Microbiology, University of Colorado Denver , Denver, CO , USA
| | - Carsten Schmitz
- Department of Biomedical Research, National Jewish Health , Denver, CO , USA ; Department of Immunology and Microbiology, University of Colorado Denver , Denver, CO , USA
| | - Anne-Laure Perraud
- Department of Biomedical Research, National Jewish Health , Denver, CO , USA ; Department of Immunology and Microbiology, University of Colorado Denver , Denver, CO , USA
| |
Collapse
|
41
|
Tian Y, Voineagu I, Paşca SP, Won H, Chandran V, Horvath S, Dolmetsch RE, Geschwind DH. Alteration in basal and depolarization induced transcriptional network in iPSC derived neurons from Timothy syndrome. Genome Med 2014; 6:75. [PMID: 25360157 PMCID: PMC4213483 DOI: 10.1186/s13073-014-0075-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/15/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Common genetic variation and rare mutations in genes encoding calcium channel subunits have pleiotropic effects on risk for multiple neuropsychiatric disorders, including autism spectrum disorder (ASD) and schizophrenia. To gain further mechanistic insights by extending previous gene expression data, we constructed co-expression networks in Timothy syndrome (TS), a monogenic condition with high penetrance for ASD, caused by mutations in the L-type calcium channel, Cav1.2. METHODS To identify patient-specific alterations in transcriptome organization, we conducted a genome-wide weighted co-expression network analysis (WGCNA) on neural progenitors and neurons from multiple lines of induced pluripotent stem cells (iPSC) derived from normal and TS (G406R in CACNA1C) individuals. We employed transcription factor binding site enrichment analysis to assess whether TS associated co-expression changes reflect calcium-dependent co-regulation. RESULTS We identified reproducible developmental and activity-dependent gene co-expression modules conserved in patient and control cell lines. By comparing cell lines from case and control subjects, we also identified co-expression modules reflecting distinct aspects of TS, including intellectual disability and ASD-related phenotypes. Moreover, by integrating co-expression with transcription factor binding analysis, we showed the TS-associated transcriptional changes were predicted to be co-regulated by calcium-dependent transcriptional regulators, including NFAT, MEF2, CREB, and FOXO, thus providing a mechanism by which altered Ca(2+) signaling in TS patients leads to the observed molecular dysregulation. CONCLUSIONS We applied WGCNA to construct co-expression networks related to neural development and depolarization in iPSC-derived neural cells from TS and control individuals for the first time. These analyses illustrate how a systems biology approach based on gene networks can yield insights into the molecular mechanisms of neural development and function, and provide clues as to the functional impact of the downstream effects of Ca(2+) signaling dysregulation on transcription.
Collapse
Affiliation(s)
- Yuan Tian
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
- />Interdepartmental Ph.D. Program in Bioinformatics, University of California, Los Angeles, CA 90095 USA
| | - Irina Voineagu
- />School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052 Australia
| | - Sergiu P Paşca
- />Department of Psychiatry & Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Hyejung Won
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Vijayendran Chandran
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Steve Horvath
- />Department of Human Genetics, David Geffen Sch. of Medicine, UCLA, Los Angeles, CA USA
| | - Ricardo E Dolmetsch
- />Department of Neurobiology, Stanford University, Stanford, CA 94305-5345 USA
- />Novartis Institutes for Biomedical Research, Cambridge, MA 02139 USA
| | - Daniel H Geschwind
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
- />Interdepartmental Ph.D. Program in Bioinformatics, University of California, Los Angeles, CA 90095 USA
- />Department of Human Genetics, David Geffen Sch. of Medicine, UCLA, Los Angeles, CA USA
| |
Collapse
|
42
|
Predicting changes in cardiac myocyte contractility during early drug discovery with in vitro assays. Toxicol Appl Pharmacol 2014; 279:87-94. [DOI: 10.1016/j.taap.2014.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/12/2014] [Accepted: 06/07/2014] [Indexed: 11/22/2022]
|
43
|
Buraei Z, Liang H, Elmslie KS. Voltage control of Ca²⁺ permeation through N-type calcium (Ca(V)2.2) channels. ACTA ACUST UNITED AC 2014; 144:207-20. [PMID: 25114024 PMCID: PMC4144670 DOI: 10.1085/jgp.201411201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Voltage dependence of permeation enhances Ca2+ influx through CaV2.2 channels relative to that of other ions at depolarized voltages. Voltage-gated calcium (CaV) channels deliver Ca2+ to trigger cellular functions ranging from cardiac muscle contraction to neurotransmitter release. The mechanism by which these channels select for Ca2+ over other cations is thought to involve multiple Ca2+-binding sites within the pore. Although the Ca2+ affinity and cation preference of these sites have been extensively investigated, the effect of voltage on these sites has not received the same attention. We used a neuronal preparation enriched for N-type calcium (CaV2.2) channels to investigate the effect of voltage on Ca2+ flux. We found that the EC50 for Ca2+ permeation increases from 13 mM at 0 mV to 240 mM at 60 mV, indicating that, during permeation, Ca2+ ions sense the electric field. These data were nicely reproduced using a three-binding-site step model. Using roscovitine to slow CaV2.2 channel deactivation, we extended these measurements to voltages <0 mV. Permeation was minimally affected at these hyperpolarized voltages, as was predicted by the model. As an independent test of voltage effects on permeation, we examined the Ca2+-Ba2+ anomalous mole fraction (MF) effect, which was both concentration and voltage dependent. However, the Ca2+-Ba2+ anomalous MF data could not be reproduced unless we added a fourth site to our model. Thus, Ca2+ permeation through CaV2.2 channels may require at least four Ca2+-binding sites. Finally, our results suggest that the high affinity of Ca2+ for the channel helps to enhance Ca2+ influx at depolarized voltages relative to other ions (e.g., Ba2+ or Na+), whereas the absence of voltage effects at negative potentials prevents Ca2+ from becoming a channel blocker. Both effects are needed to maximize Ca2+ influx over the voltages spanned by action potentials.
Collapse
Affiliation(s)
- Zafir Buraei
- Department of Biology and Health Sciences, Pace University, New York, NY 10038 Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112
| | - Haoya Liang
- Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112
| | - Keith S Elmslie
- Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112 The Baker Laboratory of Pharmacology, Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO 63501
| |
Collapse
|
44
|
Age-related homeostatic midchannel proteolysis of neuronal L-type voltage-gated Ca²⁺ channels. Neuron 2014; 82:1045-57. [PMID: 24908485 DOI: 10.1016/j.neuron.2014.04.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2014] [Indexed: 12/11/2022]
Abstract
Neural circuitry and brain activity depend critically on proper function of voltage-gated calcium channels (VGCCs), whose activity must be tightly controlled. We show that the main body of the pore-forming α1 subunit of neuronal L-type VGCCs, Cav1.2, is proteolytically cleaved, resulting in Cav1.2 fragment channels that separate but remain on the plasma membrane. This "midchannel" proteolysis is regulated by channel activity, involves the Ca(2+)-dependent protease calpain and the ubiquitin-proteasome system, and causes attenuation and biophysical alterations of VGCC currents. Recombinant Cav1.2 fragment channels mimicking the products of midchannel proteolysis do not form active channels on their own but, when properly paired, produce currents with distinct biophysical properties. Midchannel proteolysis increases dramatically with age and can be attenuated with an L-type VGCC blocker in vivo. Midchannel proteolysis represents a novel form of homeostatic negative-feedback processing of VGCCs that could profoundly affect neuronal excitability, neurotransmission, neuroprotection, and calcium signaling in physiological and disease states.
Collapse
|
45
|
Zhang-James Y, Yang L, Middleton FA, Yang L, Patak J, Faraone SV. Autism-related behavioral phenotypes in an inbred rat substrain. Behav Brain Res 2014; 269:103-14. [PMID: 24780868 DOI: 10.1016/j.bbr.2014.04.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/17/2014] [Accepted: 04/19/2014] [Indexed: 12/31/2022]
Abstract
Behavioral and genetic differences among Wistar-Kyoto (WKY) rats from different vendors and different breeders have long been observed, but generally overlooked. In our prior work, we found that two closely related WKY substrains, the WKY/NCrl and WKY/NHsd rats, differ in a small percentage of their genome which appeared to be highly enriched for autism risk genes. Although both substrains have been used widely in studies of hypertension, attention deficit/hyperactivity disorder (ADHD) and depression, they have not been tested for any autism-related behavioral phenotypes. Furthermore, these two substrains have often been used interchangeably in previous studies; no study has systematically examined the phenotypic differences that could be attributed by their small yet potentially meaningful genetic differences. In this paper we compared these two substrains on a battery of neurobehavioral tests. Although two substrains were similar in locomotor activity, WKY/NCrl rats were significantly different from WKY/NHsd rats in the elevated plus maze test, as well as measures of social interaction and ultrasonic vocalization. These strains were also compared with Sprague Dawley (SD) rats, a common outbred strain, and spontaneous hypertensive rats (SHR), an inbred rat model for ADHD and hypertension, which were derived from the same ancestor strain as the WKY strains. Our behavioral findings suggest that WKY/NCrl rats may be useful as a model autism spectrum disorders due to their lower social interest, lower ultrasonic vocalization and higher anxiety levels when WKY/NHsd rats are used as the control strain. Given the small genetic difference between the two inbred substrains, future studies to identify the exact gene and sequence variants that differ between the two may be useful for identifying the genetic mechanisms underlying these behaviors.
Collapse
Affiliation(s)
- Yanli Zhang-James
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| | - Li Yang
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.
| | - Frank A Middleton
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, United States; Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| | - Lina Yang
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| | - Jameson Patak
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| | - Stephen V Faraone
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, United States; Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| |
Collapse
|
46
|
Hasreiter J, Goldnagl L, Böhm S, Kubista H. Cav1.2 and Cav1.3 L-type calcium channels operate in a similar voltage range but show different coupling to Ca(2+)-dependent conductances in hippocampal neurons. Am J Physiol Cell Physiol 2014; 306:C1200-13. [PMID: 24760982 DOI: 10.1152/ajpcell.00329.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the central nervous system, L-type voltage-gated calcium channels (LTCCs) come in two isoforms, namely Cav1.2 and Cav1.3 channels. It has been shown previously that these channels differ in biophysical properties, in subcellular localization, and in the coupling to the gene transcription machinery. In previous work on rat hippocampal neurons we have identified an excitatory cation conductance and an inhibitory potassium conductance as important LTCC coupling partners. Notably, a stimulus-dependent interplay of LTCC-mediated Ca(2+) influx and activation of these Ca(2+)-dependent conductances was found to give rise to characteristic voltage responses. However, the contribution of Cav1.2 and Cav1.3 to these voltage responses remained unknown. Hence, the relative contribution of the LTCC isoforms therein was the focus of the current study on hippocampal neurons derived from genetically modified mice, which either lack a LTCC isoform (Cav1.3 knockout mice) or express a dihydropyridine-insensitive LTCC isoform (Cav1.2DHP(-)-knockin mice). We identified common and alternate ion channel couplings of Cav1.2 and Cav1.3 channels. Whereas hyperpolarizing Ca(2+)-dependent conductances were coupled to both Cav1.2 and Cav1.3 channels, an afterdepolarizing potential was only induced by the activity of Cav1.2 channels. Unexpectedly, the activity of Cav1.2 channels was found at relatively hyperpolarized membrane voltages. Our data add important information about the differences between Cav1.2 and Cav1.3 channels that furthers our understanding of the physiological and pathophysiological neuronal roles of these calcium channels. Moreover, our findings suggest that Cav1.3 knockout mice together with Cav1.2DHP(-)-knockin mice provide valuable models for future investigation of hippocampal LTCC-dependent afterdepolarizations.
Collapse
Affiliation(s)
- Julia Hasreiter
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lena Goldnagl
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Böhm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Breitenkamp AFS, Matthes J, Nass RD, Sinzig J, Lehmkuhl G, Nürnberg P, Herzig S. Rare mutations of CACNB2 found in autism spectrum disease-affected families alter calcium channel function. PLoS One 2014; 9:e95579. [PMID: 24752249 PMCID: PMC3994086 DOI: 10.1371/journal.pone.0095579] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 03/27/2014] [Indexed: 01/06/2023] Open
Abstract
Autism Spectrum Disorders (ASD) are complex neurodevelopmental diseases clinically defined by dysfunction of social interaction. Dysregulation of cellular calcium homeostasis might be involved in ASD pathogenesis, and genes coding for the L-type calcium channel subunits CaV1.2 (CACNA1C) and CaVβ2 (CACNB2) were recently identified as risk loci for psychiatric diseases. Here, we present three rare missense mutations of CACNB2 (G167S, S197F, and F240L) found in ASD-affected families, two of them described here for the first time (G167S and F240L). All these mutations affect highly conserved regions while being absent in a sample of ethnically matched controls. We suggest the mutations to be of physiological relevance since they modulate whole-cell Ba2+ currents through calcium channels when expressed in a recombinant system (HEK-293 cells). Two mutations displayed significantly decelerated time-dependent inactivation as well as increased sensitivity of voltage-dependent inactivation. In contrast, the third mutation (F240L) showed significantly accelerated time-dependent inactivation. By altering the kinetic parameters, the mutations are reminiscent of the CACNA1C mutation causing Timothy Syndrome, a Mendelian disease presenting with ASD. In conclusion, the results of our first-time biophysical characterization of these three rare CACNB2 missense mutations identified in ASD patients support the hypothesis that calcium channel dysfunction may contribute to autism.
Collapse
Affiliation(s)
| | - Jan Matthes
- Department of Pharmacology, University of Cologne, Cologne, Germany
| | | | - Judith Sinzig
- Department of Child and Adolescent Psychiatry and Psychotherapy, LVR-Klinik Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Gerd Lehmkuhl
- Department of Child and Adolescent Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Stefan Herzig
- Department of Pharmacology, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
48
|
Banerjee S, Riordan M, Bhat MA. Genetic aspects of autism spectrum disorders: insights from animal models. Front Cell Neurosci 2014; 8:58. [PMID: 24605088 PMCID: PMC3932417 DOI: 10.3389/fncel.2014.00058] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/07/2014] [Indexed: 01/26/2023] Open
Abstract
Autism spectrum disorders (ASDs) are a complex neurodevelopmental disorder that display a triad of core behavioral deficits including restricted interests, often accompanied by repetitive behavior, deficits in language and communication, and an inability to engage in reciprocal social interactions. ASD is among the most heritable disorders but is not a simple disorder with a singular pathology and has a rather complex etiology. It is interesting to note that perturbations in synaptic growth, development, and stability underlie a variety of neuropsychiatric disorders, including ASD, schizophrenia, epilepsy, and intellectual disability. Biological characterization of an increasing repertoire of synaptic mutants in various model organisms indicates synaptic dysfunction as causal in the pathophysiology of ASD. Our understanding of the genes and genetic pathways that contribute toward the formation, stabilization, and maintenance of functional synapses coupled with an in-depth phenotypic analysis of the cellular and behavioral characteristics is therefore essential to unraveling the pathogenesis of these disorders. In this review, we discuss the genetic aspects of ASD emphasizing on the well conserved set of genes and genetic pathways implicated in this disorder, many of which contribute to synapse assembly and maintenance across species. We also review how fundamental research using animal models is providing key insights into the various facets of human ASD.
Collapse
Affiliation(s)
- Swati Banerjee
- Department of Physiology, Center for Biomedical Neuroscience, School of Medicine, University of Texas Health Science Center San Antonio, TX, USA
| | - Maeveen Riordan
- Department of Physiology, Center for Biomedical Neuroscience, School of Medicine, University of Texas Health Science Center San Antonio, TX, USA
| | - Manzoor A Bhat
- Department of Physiology, Center for Biomedical Neuroscience, School of Medicine, University of Texas Health Science Center San Antonio, TX, USA
| |
Collapse
|
49
|
Frank CA. How voltage-gated calcium channels gate forms of homeostatic synaptic plasticity. Front Cell Neurosci 2014; 8:40. [PMID: 24592212 PMCID: PMC3924756 DOI: 10.3389/fncel.2014.00040] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/28/2014] [Indexed: 01/15/2023] Open
Abstract
Throughout life, animals face a variety of challenges such as developmental growth, the presence of toxins, or changes in temperature. Neuronal circuits and synapses respond to challenges by executing an array of neuroplasticity paradigms. Some paradigms allow neurons to up- or downregulate activity outputs, while countervailing ones ensure that outputs remain within appropriate physiological ranges. A growing body of evidence suggests that homeostatic synaptic plasticity (HSP) is critical in the latter case. Voltage-gated calcium channels gate forms of HSP. Presynaptically, the aggregate data show that when synapse activity is weakened, homeostatic signaling systems can act to correct impairments, in part by increasing calcium influx through presynaptic CaV2-type channels. Increased calcium influx is often accompanied by parallel increases in the size of active zones and the size of the readily releasable pool of presynaptic vesicles. These changes coincide with homeostatic enhancements of neurotransmitter release. Postsynaptically, there is a great deal of evidence that reduced network activity and loss of calcium influx through CaV1-type calcium channels also results in adaptive homeostatic signaling. Some adaptations drive presynaptic enhancements of vesicle pool size and turnover rate via retrograde signaling, as well as de novo insertion of postsynaptic neurotransmitter receptors. Enhanced calcium influx through CaV1 after network activation or single cell stimulation can elicit the opposite response-homeostatic depression via removal of excitatory receptors. There exist intriguing links between HSP and calcium channelopathies-such as forms of epilepsy, migraine, ataxia, and myasthenia. The episodic nature of some of these disorders suggests alternating periods of stable and unstable function. Uncovering information about how calcium channels are regulated in the context of HSP could be relevant toward understanding these and other disorders.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| |
Collapse
|
50
|
Doshi-Velez F, Ge Y, Kohane I. Comorbidity clusters in autism spectrum disorders: an electronic health record time-series analysis. Pediatrics 2014; 133:e54-63. [PMID: 24323995 PMCID: PMC3876178 DOI: 10.1542/peds.2013-0819] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE The distinct trajectories of patients with autism spectrum disorders (ASDs) have not been extensively studied, particularly regarding clinical manifestations beyond the neurobehavioral criteria from the Diagnostic and Statistical Manual of Mental Disorders. The objective of this study was to investigate the patterns of co-occurrence of medical comorbidities in ASDs. METHODS International Classification of Diseases, Ninth Revision codes from patients aged at least 15 years and a diagnosis of ASD were obtained from electronic medical records. These codes were aggregated by using phenotype-wide association studies categories and processed into 1350-dimensional vectors describing the counts of the most common categories in 6-month blocks between the ages of 0 to 15. Hierarchical clustering was used to identify subgroups with distinct courses. RESULTS Four subgroups were identified. The first was characterized by seizures (n = 120, subgroup prevalence 77.5%). The second (n = 197) was characterized by multisystem disorders including gastrointestinal disorders (prevalence 24.3%) and auditory disorders and infections (prevalence 87.8%), and the third was characterized by psychiatric disorders (n = 212, prevalence 33.0%). The last group (n = 4316) could not be further resolved. The prevalence of psychiatric disorders was uncorrelated with seizure activity (P = .17), but a significant correlation existed between gastrointestinal disorders and seizures (P < .001). The correlation results were replicated by using a second sample of 496 individuals from a different geographic region. CONCLUSIONS Three distinct patterns of medical trajectories were identified by unsupervised clustering of electronic health record diagnoses. These may point to distinct etiologies with different genetic and environmental contributions. Additional clinical and molecular characterizations will be required to further delineate these subgroups.
Collapse
Affiliation(s)
- Finale Doshi-Velez
- Center for Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, MA 02115.
| | - Yaorong Ge
- Center for Biomedical Informatics, Wake Forest University, Winston-Salem, North Carolina
| | - Isaac Kohane
- Center for Biomedical Informatics, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|