Abstract
A role for voltage-gated calcium channels (VGCCs) in psychiatric disorders has long been postulated as part of a broader involvement of intracellular calcium signalling. However, the data were inconclusive and hard to interpret. We review three areas of research that have markedly advanced the field. First, there is now robust genomic evidence that common variants in VGCC subunit genes, notably CACNA1C which encodes the L-type calcium channel (LTCC) CaV1.2 subunit, are trans-diagnostically associated with psychiatric disorders including schizophrenia and bipolar disorder. Rare variants in these genes also contribute to the risk. Second, pharmacoepidemiological evidence supports the possibility that calcium channel blockers, which target LTCCs, might have beneficial effects on the onset or course of these disorders. This is especially true for calcium channel blockers that are brain penetrant. Third, long-range sequencing is revealing the repertoire of full-length LTCC transcript isoforms. Many novel and abundant CACNA1C isoforms have been identified in human and mouse brain, including some which are enriched compared to heart or aorta, and predicted to encode channels with differing functional and pharmacological properties. These isoforms may contribute to the molecular mechanisms of genetic association to psychiatric disorders. They may also enable development of therapeutic agents that can preferentially target brain LTCC isoforms and be of potential value for psychiatric indications.
This article is part of the Special Issue on ‘L-type calcium channel mechanisms in neuropsychiatric disorders’.
1. Introduction
Several voltage-gated calcium channel (VGCC) subunit genes are found among the hundreds of loci implicating thousands of genes that have been revealed by genome-wide association studies (GWAS) of psychiatric disorders. The most notable example is CACNA1C, which encodes the L-type calcium channel (LTCC) CaV1.2 pore-forming subunit.
In many respects the GWAS signals for VGCCs are no more impressive nor transformative than most others: the effect sizes are trivial, and the causal polymorphisms, molecular mechanisms, and functional correlates of the genetic variation are unknown. However, viewed in the wider context, the demonstrated involvement of VGCC genes does have greater implications. Firstly, LTCCs and other VGCCs play a well-established role in fundamental neuronal processes relevant to psychiatric disorders, including transmitter release, synaptic plasticity, and excitation-transcription coupling (Striessnig et al., 2014; Zamponi et al., 2015; Nanou and Catterall, 2018) and impact on learning, memory, and other relevant behaviours (Wankerl et al., 2010; Kabir et al., 2017). Secondly, complementing the GWAS data, there is evidence for rare VGCC variants with penetrant effects on disease risk. Thirdly, altered intracellular calcium signalling has been documented in cells from patients with psychiatric disorders, especially bipolar disorder and depression (Dubovsky and Franks, 1983; Berridge, 2014; Harrison et al., 2021b). Complemented by emerging understanding of the structure of VGCCs (Wu et al., 2015; Tang et al., 2016), these considerations together provide a strong biological framework within which to interpret the genomic evidence. Finally, and significantly, VGCCs are druggable: calcium channel blockers (CCBs), which target the α1 subunit of LTCCs (Striessnig et al., 1998), are widely used for hypertension and other cardiovascular indications (Braunwald, 1982), whilst the gabapentinoids, which act via the α2δ subunits (Gee et al., 1996), are used for several conditions including sleep disorders, epilepsy and pain (Hong et al., 2022).
Here we summarise: (a) genomic data implicating LTCCs in psychiatric disorders; (b) epidemiological evidence for beneficial therapeutic effects of CCBs in these disorders; and (c) molecular studies revealing novel LTCC isoforms that may mediate the genetic associations and provide genomically-informed psychotropic drug targets. As outlined below, LTCCs are likely of relevance to brain processes that cut across psychiatric diagnostic boundaries. However, given that the genomic evidence has been collected using current diagnostic criteria and that the most robust associations are seen for CACNA1C, we focus here on CACNA1C’s involvement in psychosis (schizophrenia and bipolar disorder). Nevertheless, many of the issues pertain to other LTCC genes and other neuropsychiatric disease phenotypes (Heyes et al., 2015; Zamponi, 2016).
2. LTCC genes contribute to risk for many psychiatric disorders
The first genome-wide significant association between an LTCC gene and a psychiatric disorder was for CACNA1C and bipolar disorder (Sklar et al., 2008; Ferreira et al., 2008). This was followed by a number of associations between this and other VGCC loci with several psychiatric disorders, especially schizophrenia, as well as with a cross-disorder phenotype (Table 1; Cross-Disorder Group of the Psychiatric Genomics Consortium, 2013; Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium, 2018; Mullins et al., 2021; Trubetskoy et al., 2022). The latest GWAS results for schizophrenia now include significant associations to three of the four LTCC α1 subunit gene loci (Trubetskoy et al., 2022).
Table 1. Common and rare variant associations of LTCC and other VGCC genes with psychiatric disorders.
| Subunit type | Channel typea | Channel namea | Subunit name | Gene symbol | Common variantsb,c | Rare variantsb,d |
|---|---|---|---|---|---|---|
| Alpha1 (α1) | L-type | Cav1.1 | α1S | CACNA1S | Scz, XD | Scz |
| Cav1.2 | α1C | CACNA1C | BD, Scz, ASD, XD | ASD, BD, Scz | ||
| Cav1.3 | α1D | CACNA1D | Scz, XD | ASD, BD | ||
| C3v1.4 | α1F | CACNA1F | ||||
| P/Q-type | Cav2.1 | α1A | CACNA1A | |||
| N-type | Cav2.2 | α1B | CACNA1B | BD | BD, Scz | |
| R-type | Cav2.3 | α1E | CACNA1E | MDD, XD | ||
| T-type | Cav3.1 | α1G | CACNA1G | Scz | ||
| Cav3.2 | α1H | CACNA1H | ASD, Scz | |||
| Cav3.3 | α1I | CACNA1I | Scz, ASD | |||
| Beta (β) | β1 | CACNB1 | ||||
| β2 | CACNB2 | Scz, BD, XD | ASD | |||
| β3 | CACNB3 | |||||
| β4 | CACNB4 | Scz | ||||
| Alpha2delta (α2δ) | α2δ1 | CACNA2D1 | MDD | Scz | ||
| α2δ2 | CACNA2D2 | Scz, XD | Scz | |||
| α2δ3 | CACNA2D3 | ASD | ||||
| α2δ4 | CACNA2D4 | XD | Scz |
Channel type and name are defined by the α1 subunit.
ADHD: attention-deficity hyperactivity disorder; ASD: autistic spectrum disorder. BD: bipolar disorder. MDD: major depression. Scz: schizophrenia. XD: cross-disorder (scz/BD/MDD/ADHD/ASD).
Common variant locus associations reported in one or more GWAS. Results in boldface are genome-wide significant in the latest Psychiatric Genomics Consortium analyses of schizophrenia (Trubetskoy et al., 2022) and bipolar disorder (Mullins et al., 2021).
Boldface denotes significant findings in the SCHEMA whole exome study of schizophrenia (Singh et al., 2022).
The cumulative GWAS data strongly suggest a role for common variation in VGCC subunit genes, especially CACNA1C, in the genetic architecture of severe mental illness (Casamassima et al., 2010; Bhat et al., 2012; Harrison et al., 2021b). However, as with all GWAS findings, many questions remain to be addressed, beyond the basic limitation that the causal gene(s) and variant(s) at each locus need to be determined (Harrison, 2015; Wang et al., 2019; Mountjoy et al., 2021; Wainberg et al., 2022). Firstly, what is the molecular mechanism of the genetic association? The genomic signals are non-coding, and so likely impact on gene regulation and expression. However, studies to date are inconclusive as to the direction of effect, if any, of the risk alleles on CACNA1C mRNA abundance (Bigos et al., 2010; Gershon et al., 2014; Yoshimizu et al., 2015; Jaffe et al., 2020); the issue is relevant since altered Cacna1c expression in rodents affects their behavioural phenotype (see Moon et al., 2018). The variable results may reflect temporal or spatial variation of the allelic effect, for example across neurodevelopment or between brain regions or cell types. Alternatively, given that the GWAS signal arises from an intron within the gene, a plausible mechanism of association is that it involves altered splicing and thence production of specific isoforms rather than an overall increase or decrease in CACNA1C expression. This possibility has yet to be tested, but a genotype effect on splicing is a key event for other psychosis risk genes (Kleinman et al., 2011; Tao et al., 2014; Xiao et al., 2017; Gandal et al., 2018; Zhang et al., 2022). Secondly, what is the downstream effect of the disease-associated genetic variation on the properties of the encoded channel and thence the processes and networks in which they participate? There are some findings showing CACNA1C genotype influences channel characteristics (Yoshimizu et al., 2015; Birey et al., 2017), and similarly for CACNA1I (Baez-Nietro et al., 2022), but the overall picture remains unclear, in part because the existence of any genotype-associated isoform(s) has yet to be shown. It is also unclear how the LTCC findings relate to the dysregulation of intracellular calcium signalling observed in several psychiatric disorders noted above, especially since much of that work was carried out in platelets and lymphocytes. However, the increasing evidence for expression and function of LTCCs in non-excitable cells indicates that a link of some kind is possible (Alves et al., 2019; Pitt et al., 2021). Thirdly, the pleiotropic nature of genetic associations across disorders raises the question as to which aspects of the clinical phenotype, as well as the pathophysiology, the LTCCs participate in (Lee et al., 2021). A parsimonious explanation is that they contribute to the processes underlying one or more of the many and diverse features observed trans--diagnostically across the disorders, such as cognitive impairment, mood instability, sleep difficulties, or physical comorbidity. The temporal profile of LTCC expression in brain across development also needs to be borne in mind when considering how, when, and where the genes exert their influence (Dedic et al., 2018; Clifton et al., 2021; Hall and Bray, 2022). Clarifying these various genotype-phenotype relationships will be one of the issues to be resolved before the role of LTCCs in disease pathogenesis can be understood, and any potential of LTCCs as psychotropic drug targets can be realized.
Complementing the common variation, rare variants in some LTCCs, and other VGCCs, also confer risk for psychiatric disorders and neurodevelopmental syndromes (Table 1). The paradigm example is Timothy syndrome, in which autistic features are prominent together with a cardiac and skeletal phenotype, caused by a gain-of-function mutation in CACNA1C (Splawski et al., 2004) which acts, at least in part, by altering splicing of the gene (Panagiotakos et al., 2019). Rare (Purcell et al., 2014; Wang et al., 2022) and structural (Song et al., 2018) variants in CACNA1C have also been reported in psychosis, and rare variants in CACNA1C and CACNA1D identified in other neurodevelopmental syndromes (Pinggera et al., 2015; Ortner et al., 2020; Rodan et al., 2021). Although of limited population impact, rare variants are invaluable because they provide greater traction on the underlying biology, and the findings may give clues as to the mechanisms underlying the common variant associations.
In summary, there is now strong evidence that LTCCs are part of the genetic architecture of a range of psychiatric disorders. Attention can now turn to understanding the nature and mechanisms of the LTCC contribution to aetiology, and whether and how this understanding can be harnessed to advance the candidacy of these channels as targets to treat psychiatric disorders.
3. Calcium channels and blockers in psychiatry
Initial interest in the possible role of LTCCs in the treatment of psychiatric disorders was stimulated by the introduction of the first calcium channel blockers (CCBs), verapamil and diltiazem, for treatment of hypertension and other cardiovascular indications. This chimed with a hypothesized role of aberrant calcium signalling in bipolar disorder and depression that was emerging around that time (Crammer, 1977; Jimerson et al., 1979; Dubovsky and Franks, 1983; Bowden et al., 1988), and with the fact that some antipsychotic drugs were shown to be calcium channel antagonists, potentially contributing to their mode of action (Gould et al., 1983). A number of case reports, case series, and small clinical trials followed, in which patients with bipolar disorder, depression or schizophrenia were treated with CCBs, mainly verapamil. Despite initial enthusiasm, findings were not robust and interest waned (Hollister and Trevino, 1999), and a systematic review in bipolar disorder concluded that there was an absence of evidence from randomized controlled trials to support their use (Cipriani et al., 2016).
Discovery that LTCCs are part of the genetic risk architecture for neuropsychiatric disorders has rekindled interest in these channels as therapeutic targets (Dubovsky, 2019; Harrison et al., 2020b). Contemporary therapeutic investigations are using the newer dihydropyridine CCBs (Ostacher et al., 2014; Atkinson et al., 2019; Burdick et al., 2020; Vahdani et al., 2020). For example, Ostacher et al. (2014) studied effects of isradipine on bipolar disorder, selecting patients based on CACNA1C risk genotype. However, these recent studies are all exploratory or pilot in nature, and have not yielded any conclusive results. While larger clinical trials are awaited, two further approaches are being adopted to advance the candidacy of CCBs in psychiatry: the first is to use pharmacoepidemiology to provide clues as to whether the existing CCBs are associated with differences in risk for, or outcome of, psychiatric disorders. The second approach is to explore the identity and characteristics of LTCCs and their subunits in more detail, with particular regard to the possibility that they may differ in the brain compared to peripheral tissues. These issues are relevant both for explaining the mechanisms by which the genes may contribute to risk for psychiatric disorders, and for the possibility that modified CCBs – or other therapeutic agents interacting with these channels in order to enhance, inhibit, or modulate their functioning – could be of value.
3.1. Pharmacoepidemiology
The advent of electronic health records has enabled large-scale pharmacoepidemiological studies of patients who are prescribed CCBs for cardiovascular indications to study whether their use is associated with an altered incidence or course of psychiatric disorders. A study of the Swedish population found that patients with schizophrenia or bipolar disorder had fewer psychiatric hospital admissions when they were taking CCBs compared to when they were not (Hayes et al., 2019). Other studies have compared people taking CCBs with people taking another class of antihypertensive drug, to control for the confounding effect of hypertension. Being conducted more recently than the initial clinical studies mentioned above, the pharmacoepidemiological work focuses on the dihydropyridine CCBs which have largely superseded use of verapamil and diltiazem; of the dihydropyridines, amlodipine is much the most frequently prescribed.
The pharmacoepidemiological findings are somewhat varied, but overall there is evidence for a reduced risk of onset or recurrence of depression with CCBs compared to beta-blockers, and some evidence for CCBs compared to diuretics; in contrast, CCBs are inferior compared to angiotensin II receptor blockers (Boal et al., 2016; Cao et al., 2019; Kessing et al., 2019; Agustini et al., 2020; Colbourne et al., 2021; Shaw et al., 2021). A broadly similar pattern for CCBs relative to other anti-hypertensive classes pertains for several other psychiatric disorders (Colbourne et al., 2021), delirium (Harrison et al., 2020a), and neurodegenerative disorders (see Harrison et al., 2021a).
An important caveat is that these pharmacoepidemiological studies considered CCBs as a class, and did not take the varying blood-brain barrier penetrability of individual drugs into account. It seems likely that any psychiatric benefits of CCBs would be greater for those drugs that do enter the brain and thereby have the opportunity to block neuronal LTCCs compared to those that do not. Notably, three of the CCBs mentioned thus far – verapamil, diltiazem, and amlodipine – all have low blood-brain barrier penetrability unlike the other dihydropyridines (such as felodipine, isradipine and nifedipine). We tested this hypothesis in a retrospective cohort study using a large electronic health records network (Colbourne and Harrison, 2022). We found that, over a two-year exposure period, the brain-penetrant drugs were associated with a lower incidence of a first psychiatric diagnosis compared to the other CCBs. This applied across a range of common disorders, as well as for delirium and dementia, with an overall risk reduction of 12% (Fig. 1). Small reductions in risk were also seen for recurrence of prior diagnoses. In addition, the brain-penetrant CCBs were associated with a lower overall incidence of psychiatric disorders compared to the angiotensin receptor blockers. This in contrast to the earlier finding that angiotensin II receptor blockers were associated with a lower incidence when compared to all CCBs (of which the large majority of prescriptions were for non-penetrant CCBs).
Fig. 1. Brain-penetrant calcium channel blockers and risk of psychiatric disorders.
The incidence of a first recorded psychiatric diagnosis during a two-year period is lower in patients taking a brain-penetrant CCB than those taking amlodipine, a CCB with low brain permeability (n = 44,732 in each cohort). Cohorts were propensity score matched at baseline for age, sex, race, blood pressure, body mass index, and for a range of medical diagnoses and medications. Risk ratios are shown with 95% confidence intervals. Similar results were seen when brain-penetrant CCBs were compared with verapamil and diltiazem. Data taken from Colbourne and Harrison (2022).
Pharmacoepidemiological studies are observational, subject to residual confounding, and cannot demonstrate causality nor mechanism. Nevertheless, the results suggest that brain-penetrant CCBs may have some relative benefits on the risk of common psychiatric disorders. As such the findings provide impetus to explore further the role of brain LTCCs in these disorders and as treatment targets for novel psychotropic drugs. Notwithstanding this rationale, the existing CCBs, even those that readily access the brain, are likely not optimal at occupying central LTCCs, and even if this could be achieved at higher doses, it would come at the price of significant side-effects due to their cardiovascular actions. A more attractive option would be to modify CCBs to enable them to preferentially block brain VGCCs.
3.2. Towards brain-selective LTCC drugs
The potential to selectively target brain LTCCs is afforded by the presence of brain-enriched isoforms. (Note that the term ‘isoform’ is sometimes used to refer to LTCC subtypes, e.g. CaV1.2 vs CaV1.3, but here we use it in its formal sense to refer to alternative mRNA transcripts and proteins expressed from a single gene). It has been known for a while that alternative splicing of specific exons within individual LTCC genes is an important mechanism contributing to the functional diversity of the resulting channels (Lipscombe et al., 2013; Lipscombe and Andrade, 2015). CACNA1C contains many alternatively spliced exons, including exons 8/8A, involved in Timothy syndrome, and exons 21/22, 31/31a, and 33. These exons have been shown in various ways to impact upon channel kinetics, state-dependent inactivation, and dihydropyridine binding (Soldatov et al., 1995; Zuhlke et al., 1998; Liao et al., 2007; Tiwari et al., 2006; Hu et al., 2017; Li et al., 2017). These splicing events can be developmentally regulated and tissue-specific, including some exons that are retained in brain (e.g. exon 22; Tang et al., 2007).
Recently, application of long-range cDNA sequencing has revealed many new splicing events in LTCCs and enabled identification of the repertoire of full-length transcripts within which alternative exons occur (Clark et al., 2020). Prior to these technical advances, methods primarily focused on individual exons or segments of the gene, and could not readily detect previously unannotated exons. The long-read data highlight that pre-existing transcriptome annotations for CACNA1C are far from complete, and give a misleading picture (Clark et al., 2020; Hall et al., 2021). Importantly, several of the newly identified human CAC-NA1C isoforms are more abundant than the annotated isoforms, and are enriched in brain compared to heart or aorta (NALH, SMH, HL, PJH, WH, EMT, unpublished observations). Moreover, some of the new isoforms are predicted to encode channels that vary in their function and/or pharmacology. The isoforms arise from multiple sources, including alternative transcription start sites, inclusion of novel exons, and different combinations of known exons. There is also heterogeneity at the 3′-end encoding the carboxy terminal domain. Some of the novel isoforms are common to humans and mice, whereas others differ between these species (SMH, NALH, PJH, WH, EMT, unpublished observations).
Many questions remain regarding the significance of these observations. It is not known which of the novel transcripts are translated and whether transcript abundance predicts protein abundance. Nor is it known where they are expressed (in terms of cell type and subcellular location), nor how they are regulated. It also remains to be determined whether the novel isoforms contribute to the mechanism by which genetic variants in CACNA1C influence risk for psychiatric disorders. Furthermore, the predicted functional and pharmacological differences between the predominant isoforms in brain and periphery need to be empirically tested using a range of methods. Given the species differences mentioned, the approaches will need to include human model systems, such as induced pluripotent stem cell-derived neurons and other relevant cell types (De Los Angeles et al., 2021). Finally, it is important to bear in mind that even an optimal centrally-acting dihydropyridine may not achieve full blockade due to the membrane potential of neurons (Xu and Lipscombe, 2001).
Notwithstanding these important caveats, the recent molecular findings provide a tantalizing possibility that brain-enriched isoforms of CACNA1C - and other VGCCs - could be preferentially targeted, allowing therapeutic agents for psychiatric disorders with greater central potency and fewer peripheral side-effects (Harrison et al., 2020b; Hall and Tunbridge, 2021). Certainly, their existence complements the opportunities, and difficulties, that exist in terms of identifying drugs that can target one LTCC selectively from another, such as CaV1.2 vs. CaV1.3 (Zamponi et al., 2015; Ortner et al., 2017; Wang et al., 2018; Lanzetti and Di Biase, 2022).
4. Conclusions
The diverse and cumulatively compelling evidence outlined above suggests that LTCCs, and some other VGCCs, contribute to the mechanisms underlying serious mental illnesses. Compared to most of the genes linked to these disorders, considerably more is known about the function and structure of the encoded proteins. Critically, CCBs and gabapentinoids show that LTCCs are druggable and, moreover, that these drugs can impact on psychiatric phenotypes. These factors together give LTCCs a high priority in the efforts to leverage genomic discoveries to clarify the pathophysiology and advance the pharmacotherapy of serious psychiatric disorders (Breen et al., 2016; Birnbaum and Weinberger, 2020; Harrison et al., 2022). Detailed functional characterization of LTCC gene isoforms that are preferentially expressed in brain, and those that are modulated by psychiatric risk genotype, will be a key part of this endeavour.
Acknowledgments
Supported by the National Institute for Health and Care Research (NIHR) Oxford Health Biomedical Research Centre, and an unrestricted grant from J&J Innovations to EMT and PJH. SH is part funded by a Marriott Scholarship from Wolfson College, Oxford. LC is a Wellcome Trust Clinical Doctoral Fellow. WH is supported by the Biotechnology and Biological Sciences Research Council (BBSRC). The views expressed are those of the authors and not necessarily those of the National Health Service, NIHR, or the Department of Health and Social Care.
Footnotes
Author contributions
Harrison – conceived idea and wrote manuscript. Husain, Lee, De Los Angeles, Colbourne, Mould, Hall, Haerty, Tunbridge – critically reviewed and edited manuscript. All authors approved submission.
Data availability
No data was used for the research described in the article.
References
- Agustini B, Mohebbi M, Woods RL, McNeil JJ, Nelson MR, Shah RC, Murray AM, Ernst ME, Reid CM, Tonkin A, Lockery JE, et al. on behalf of the ASPREE Investigator Group The association of antihypertensive use and depressive symptoms in large older population with hypertension living in Australia and the United States: a cross-sectional study. J Hum Hypertens. 2020;34:787–794. doi: 10.1038/s41371-020-0303-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alves VS, Alves-Silva HS, Orts DJB, Ribeiro-Silva L, Ariciso-Miranda M, Oliveria FA. Calcium signaling in neurons and glial cells: role of Cav1 channels. Neuroscience. 2019;421:95–111. doi: 10.1016/j.neuroscience.2019.09.041. [DOI] [PubMed] [Google Scholar]
- Atkinson LZ, Colbourne L, Smith A, Harmer CH, Nobre AC, Rendell J, Jones H, Hinds C, Mould A, Tunbridge EM, Cipriani A, et al. The Oxford study of Calcium channel Antagonism, Cognition, Mood instability and Sleep (OxCaMS): study protocol for a randomised controlled, experimental medicine study. Trials. 2019;20:120. doi: 10.1186/s13063-019-3175-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baez-Nietro D, Allen A, Akers-Campbell S, Yang L, Budnik N, Pupo A, Shin YC, Genovese G, Liao M, Perez-Palma E, Heyne H, et al. Analysing an allelic series of rare missense variants of CACNA1I in a Swedish schizophrenia cohort. Brain. 2022;145:1839–1855. doi: 10.1093/brain/awab443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berridge MJ. Calcium signalling and psychiatric disease: bipolar disorder and schizophrenia. Cell Tissue Res. 2014;357:477–492. doi: 10.1007/s00441-014-1806-z. [DOI] [PubMed] [Google Scholar]
- Bhat S, Dao DT, Terrillion CE, Arad M, Smith RJ, Soldatov NM, Gould TD. CACNA1C (Cav1.2) in the pathophysiology of psychiatric disease. Prog Neurobiol. 2012;99:1–14. doi: 10.1016/j.pneurobio.2012.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bigos KL, Mattay VS, Callicott JH, Straub RE, Vakkalanka R, Kolachana B, Hyde TM, Lipska BK, Kleinman JE, Weinberger DR. Genetic variation in CACNA1C affects brain circuitries related to mental illness. Arch Gen Psychiatr. 2010;67:939–945. doi: 10.1001/archgenpsychiatry.2010.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium. Genomic dissection of bipolar disorder and schizophrenia, including 28 subphenotypes. Cell. 2018;173:1705–1715.:e16. doi: 10.1016/j.cell.2018.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birey F, Andersen J, Makinson CD, Islam S, Wei W, Huber N, Fan HC, Metzler KRC, Panagiotakos G, Thom N, O’Rourke NA, et al. Assembly of functionally integrated human forebrain spheroids. Nature. 2017;545:54–59. doi: 10.1038/nature22330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birnbaum R, Weinberger DR. Pharmacological implications of emerging schizophrenia genetics: can the bridge from ‘genomics’ to ‘therapeutics’ be defined and traversed? J Clin Psychopharmacol. 2020;40:323–329. doi: 10.1097/JCP.0000000000001215. [DOI] [PubMed] [Google Scholar]
- Boal AH, Smith DJ, McCallum L, Muir S, Touyz RM, Dominiczak AF, Padmanabhan S. Monotherapy with major antihypertensive drug classes and risk of hospital admissions for mood disorders. Hypertension. 2016;68:1132–1138. doi: 10.1161/HYPERTENSIONAHA.116.08188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowden CL, Huang LG, Javors MA, Johnson JM, Seleshi E, McIntyre K, Contreras S, Maas JW. Calcium function in affective disorders and healthy controls. Biol Psychiatr. 1988;23:367–376. doi: 10.1016/0006-3223(88)90287-9. [DOI] [PubMed] [Google Scholar]
- Braunwald E. Mechanism of action of calcium-channel-blocking agents. N Engl J Med. 1982;307:1618–1627. doi: 10.1056/NEJM198212233072605. [DOI] [PubMed] [Google Scholar]
- Breen G, Li Q, Roth BL, O’Donnell P, Didriksen M, Dolmetsch R, O’Reilly PF, Gaspar HA, Manji H, Huebel C, Kelsoe JR, et al. Translating genome-wide association findings into new therapeutics for psychiatry. Nat Neurosci. 2016;19:1392–1396. doi: 10.1038/nn.4411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burdick KE, Perez-Rodriguez M, Birnbaum R, Shanahan M, Larsen E, Harper C, Poskus J, Sklar P. A molecular approach to treating cognition in schizophrenia by calcium channel blockade: an open-label pilot study of the calcium-channel antagonist isradipine. Schizophr Res Cogn. 2020;21:100180. doi: 10.1016/j.scog.2020.100180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao YY, Xiang X, Song J, Tian YH, Wang MY, Wang XW, Li M, Huang Z, Wu Y, Wu T, Wu WQ, et al. Distinct effects of antihypertensives on depression in the real-world setting: a retrospective cohort study. J Affect Disord. 2019;259:386–391. doi: 10.1016/j.jad.2019.08.075. [DOI] [PubMed] [Google Scholar]
- Casamassima F, Hay AC, Benedetti A, Lattanzi L, Cassano GB, Perlis RH. L-type calcium channels and psychiatric disorders: a brief review. Am J Med Genet. 2010;153B:1373–1390. doi: 10.1002/ajmg.b.31122. [DOI] [PubMed] [Google Scholar]
- Cipriani A, Saunders K, Attenburrow MJ, Stefaniak J, Panchal P, Stockton S, Lane TA, Tunbridge EM, Geddes JR, Harrison PJ. A systematic review of calcium channel antagonists in bipolar disorder and some considerations for their future development. Mol Psychiatr. 2016;21:1324–1332. doi: 10.1038/mp.2016.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clark MB, Wrzesinski T, Garcia AB, Hall NAL, Kleinman JE, Hyde T, Weinberger DR, Harrison PJ, Haerty W, Tunbridge EM. Long-read sequencing reveals the complex splicing profile of the psychiatric risk gene CACNA1C in human brain. Mol Psychiatr. 2020;25:37–47. doi: 10.1038/s41380-019-0583-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clifton NE, Collado-Torres L, Burke EE, Pardinas AF, Harwood JC, Di Florio A, Walters JTR, Owen MJ, O’Donovan MC, Weinberger DR, Holmans PA, et al. Developmental profile of psychiatric risk associated with voltage-gated cation channel activity. Biol Psychiatr. 2021;90:399–408. doi: 10.1016/j.biopsych.2021.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colbourne L, Harrison PJ. Brain-penetrant calcium channel blockers are associated with a reduced incidence of neuropsychiatric disorders. Mol Psychiatr. 2022 doi: 10.1038/s41380-022-01615-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colbourne L, Luciano S, Harrison PJ. Onset and recurrence of psychiatric disorders associated with anti-hypertensive drug classes. Transl Psychiatry. 2021;11:319. doi: 10.1038/s41398-021-01444-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crammer JL. Calcium metabolism and mental disorder. Psychol Med. 1977;7:557–560. doi: 10.1017/s003329170000619x. [DOI] [PubMed] [Google Scholar]
- Cross-Disorder Group of the Psychiatric Genomics Consortium. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet. 2013;381:1371–1379. doi: 10.1016/S0140-6736(12)62129-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Los Angeles A, Brennand KJ, Hall NA, Fernando M, Harrison PJ, Maher BJ, Weinberger DR, Tunbridge EM. Induced pluripotent stem cells in psychiatry: an overview and critical perspective. Biol Psychiatr. 2021;90:362–372. doi: 10.1016/j.biopsych.2021.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dedic N, Pohlmann ML, Richter JS, Mehta D, Czamara D, Metzger MW, Dine J, Bedenk BT, Hartmann J, Wagner KV, Jurik A, et al. Cross-disorder risk gene CACNA1C differentially modulates susceptibility to psychiatric disorders during development and adulthood. Mol Psychiatr. 2018;23:533–543. doi: 10.1038/mp.2017.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dubovsky SL. Applications of calcium channel blockers in psychiatry: pharmacokinetic and pharmacodynamic aspects of treatment of bipolar disorder. Expet Opin Drug Metabol Toxicol. 2019;15:35–47. doi: 10.1080/17425255.2019.1558206. [DOI] [PubMed] [Google Scholar]
- Dubovsky SL, Franks RD. Intracellular calcium ions in affective disorders: a review and an hypothesis. Biol Psychiatr. 1983;18:781–797. [PubMed] [Google Scholar]
- Ferreira MA, O’Donovan MC, Meng YA, Jones IR, Ruderfer DM, Jones L, Fan J, Kirov G, Perlis RH, Green EK, Smoller JW, et al. Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nat Genet. 2008;40:1056–1058. doi: 10.1038/ng.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gandal MJ, Zhang P, Hadjimichael E, Walker RL, Chen C, Liu S, Won H, van Bakel H, Varghese M, Wang Y, Shieh AW, et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science. 2018;362 doi: 10.1126/science.aat8127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gee NS, Brown JP, Dissanayake VUK, Offord J, Thurlow R, Woodruff GN. The novel anticonvulsant drug, gabapentin (Neurontin) binds to the alpha2δ subunit of a calcium channel. J Biol Chem. 1996;271:5768–5776. doi: 10.1074/jbc.271.10.5768. [DOI] [PubMed] [Google Scholar]
- Gershon ES, Grennan K, Busnello J, Badner JA, Ovsiew F, Memon S, Alliey-Rodriguez N, Cooper J, Romanos B, Liu C. A rare mutation of CACNA1C in a patient with bipolar disorder, and decreased gene expression associated with a bipolar-associated common SNP of CACNA1C in brain. Mol Psychiatr. 2014;19:890–894. doi: 10.1038/mp.2013.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gould RJ, Murphy KM, Reynolds IJ, Snyder SH. Antischizophrenic drugs of the diphenylbutylpiperidine type act as calcium channel antagonists. Proc Natl Acad Sci U S A. 1983;80:5122–5125. doi: 10.1073/pnas.80.16.5122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall J, Bray NJ. Schizophrenia genomics: convergence on synaptic development, adult synaptic plasticity, or both? Biol. Psychiatr. 2022;91:709–717. doi: 10.1016/j.biopsych.2021.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall NAL, Tunbridge EM. Brain-enriched CACNA1C isoforms as novel, selective targets for psychiatric indications. Neuropsychopharmacology. 2021 doi: 10.1038/s41386-021-01114-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall NAL, Carlyle BC, Haerty W, Tunbridge EM. Roadblock: improved annotations do not necessarily translate into new functional insights. Genome Biol. 2021;22:320. doi: 10.1186/s13059-021-02542-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ. Recent genetic findings in schizophrenia and their therapeutic relevance. J Psychopharmacol. 2015;29:85–96. doi: 10.1177/0269881114553647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ, Luciano S, Colbourne L. Rates of delirium associated with calcium channel blockers compared to diuretics, renin-angiotensin system agents and beta-blockers: an electronic health records network study. J Psychopharmacol. 2020a;34:848–855. doi: 10.1177/0269881120936501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ, Tunbridge EM, Dolphin AC, Hall J. Voltage-gated calcium channel blockers for psychiatric disorders: genomic reappraisal. Br J Psychiatry. 2020b;216:250–253. doi: 10.1192/bjp.2019.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ, Colbourne L, Luciano S. Incidence of neurodegenerative and cerebrovascular diseases associated with antihypertensive drug classes. Br J Psychiatry. 2021a:1–3. doi: 10.1192/bjp.2020.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ, Hall N, Mould A, Al-Juffali N, Tunbridge EM. Cellular calcium in bipolar disorder: systematic review and meta-analysis. Mol Psychiatr. 2021b;26:4106–4116. doi: 10.1038/s41380-019-0622-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrison PJ, Mould A, Tunbridge EM. New drug targets in psychiatry: neurobiological considerations in the genomics era. Neurosci Biobehav Rev. 2022;139:104763. doi: 10.1016/j.neubiorev.2022.104763. [DOI] [PubMed] [Google Scholar]
- Hayes JF, Lundin A, Wicks S, Lewis G, Wong ICK, Osborn DPJ, Dalman C. Association of hydroxylmethyl glutaryl coenzyme A reductase inhibitors, L-Type calcium channel antagonists, and biguanides with rates of psychiatric hospitalization and self-harm in individuals with serious mental illness. JAMA Psychiatr. 2019;76:382–390. doi: 10.1001/jamapsychiatry.2018.3907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heyes S, Pratt WS, Rees E, Dahimene S, Ferron L, Owen MJ, Dolphin AC. Genetic disruption of voltage-gated calcium channels in psychiatric and neurological disorders. Prog Neurobiol. 2015;134:36–54. doi: 10.1016/j.pneurobio.2015.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hollister LE, Trevino ES. Calcium channel blockers in psychiatric disorders: a review of the literature. Can J Psychiatr. 1999;44:658–664. doi: 10.1177/070674379904400702. [DOI] [PubMed] [Google Scholar]
- Hong JSW, Atkinson LZ, Al-Juffali N, Awad A, Geddes JR, Tunbridge EM, Harrison PJ, Cipriani A. Gabapentin and pregabalin in bipolar disorder, anxiety states, and insomnia: systematic review, meta-analysis, and rationale. Mol Psychiatr. 2022;27:1339–1349. doi: 10.1038/s41380-021-01386-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Z, Liang MC, Soong TW. Alternative splicing of L-type CaV1.2 calcium channels: implications in cardiovascular diseases. Genes. 2017;8 doi: 10.3390/genes8120344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaffe AE, Hoeppner DJ, Saito T, Blanpain L, Ukaigwe J, Burke EE, Collado-Torres L, Tao R, Tajinda K, Maynard KR, Tran MN, et al. Profiling gene expression in the human dentate gyrus granule cell layer reveals insights into schizophrenia and its genetic risk. Nat Neurosci. 2020;23:510–519. doi: 10.1038/s41593-020-0604-z. [DOI] [PubMed] [Google Scholar]
- Jimerson DC, Post RM, Carman JS, van Kammen DP, Wood JH, Goodwin FK, Bunney WE., Jr CSF calcium: clinical correlates in affective illness and schizophrenia. Biol Psychiatr. 1979;14:37–51. [PubMed] [Google Scholar]
- Kabir ZD, Martinez-Rivera A, Rajadhyaksha AM. From gene to behavior: L-Type calcium channel mechanisms underlying neuropsychiatric symptoms. Neurotherapeutics. 2017;14:588–613. doi: 10.1007/s13311-017-0532-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kessing LV, Rytgaard HC, Gerds TA, Berk M, Ekstrom CT, Andersen PK. New drug candidates for depression - a nationwide population-based study. Acta Psychiatr Scand. 2019;139:68–77. doi: 10.1111/acps.12957. [DOI] [PubMed] [Google Scholar]
- Kleinman JE, Law AJ, Lipska BK, Hyde TM, Ellis JK, Harrison PJ, Weinberger DR. Genetic neuropathology of schizophrenia: new approaches to an old question and new uses for postmortem human brains. Biol Psychiatr. 2011;69:140–145. doi: 10.1016/j.biopsych.2010.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lanzetti S, Di Biase V. Small molecules as modulators of voltage-gated calcium channels in neurological disorders: state of the art and perspectives. Molecules. 2022;27:1312. doi: 10.3390/molecules27041312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee PH, Feng YA, Smoller JW. Pleiotropy and cross-disorder genetics among psychiatric disorders. Biol Psychiatr. 2021;89:20–31. doi: 10.1016/j.biopsych.2020.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G, Wang J, Liao P, Bartels P, Zhang H, Yu D, Liang MC, Poh KK, Yu CY, Jiang F, Yong TF, et al. Exclusion of alternative exon 33 of CaV1.2 calcium channels is proarrythmogenic. Proc Natl Acad Sci USA. 2017;114:E4288–E4295. doi: 10.1073/pnas.1617205114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liao P, Yu D, Yong TF, Soon JL, Chua YL, Soong TW. A smooth muscle CaV1.2 calcium channel splice variant underlies hyperpolarized window current and enhanced state-dependent inhibition by nifedipine. J Biol Chem. 2007;282:35133–35142. doi: 10.1074/jbc.M705478200. [DOI] [PubMed] [Google Scholar]
- Lipscombe D, Andrade A. Calcium channel CaValpha(1) splice isoforms - tissue specificity and drug action. Curr Mol Pharmacol. 2015;8:22–31. doi: 10.2174/1874467208666150507103215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipscombe D, Andrade A, Allen SE. Alternative splicing: functional diversity among voltage-gated calcium channels and behavioral consequences. Biochim Biophys Acta. 2013;1828:1522–1529. doi: 10.1016/j.bbamem.2012.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moon AL, Haan N, Wilkinson LS, Thomas KL, Hall J. CACNA1C: association with psychiatric disorders, behaviour, and neurogenesis. Schizophr Bull. 2018;44:958–965. doi: 10.1093/schbul/sby096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mountjoy E, Schmidt EM, Carmona M, Schwartzentruber J, Peat G, Miranda A, Fumis L, Hayhurst J, Buniello A, Karim MA, Wright D, et al. An open approach to systematically prioritize causal variants and genes at all published human GWAS trait-associated loci. Nat Genet. 2021;53:1527–1533. doi: 10.1038/s41588-021-00945-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mullins N, Forstner AJ, O’Connell KS, Coombes B, Coleman JRI, Qiao Z, Als TD, Bigdeli TB, Borte S, Bryois J, Charney AW, et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat Genet. 2021;53:817–829. doi: 10.1038/s41588-021-00857-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nanou E, Catterall WA. Calcium channels, synaptic plasticity, and neuropsychiatric disease. Neuron. 2018;98:466–481. doi: 10.1016/j.neuron.2018.03.017. [DOI] [PubMed] [Google Scholar]
- Ortner NJ, Bock G, Dougalis A, Kharitonova M, Duda J, Hess S, Tuluc P, Pomberger T, Stefanova N, Pitterl F, Ciossek T, et al. Lower affinity of isradipine for L-Type Ca(2 +) channels during substantia nigra dopamine neuron-like activity: implications for neuroprotection in Parkinson’s disease. J Neurosci. 2017;37:6761–6777. doi: 10.1523/JNEUROSCI.2946-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ortner NJ, Kaserer T, Copeland JN, Striessnig J. De novo CACNA1D Ca(2 +) channelopathies: clinical phenotypes and molecular mechanisms. Pflügers Archiv. 2020;472:755–773. doi: 10.1007/s00424-020-02436-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ostacher MJ, Iosifescu DV, Hay A, Blumenthal SR, Sklar P, Perlis RH. Pilot investigation of isradipine in the treatment of bipolar depression motivated by genome-wide association. Bipolar Disord. 2014;16:199–203. doi: 10.1111/bdi.12143. [DOI] [PubMed] [Google Scholar]
- Panagiotakos G, Haveles C, Arjun A, Petrova R, Rna A, Portmann T, Pasca SP, Palmer TD, Dolmetsch RE. Aberrant calcium channel splicing drives defects in cortical differentiation in Timothy syndrome. Elife. 2019;8:e51037. doi: 10.7554/eLife.51037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinggera A, Lieb A, Benedetti B, Lampert M, Monteleone S, Liedl KR, Tuluc P, Striessnig J. CACNA1D de novo mutations in autism spectrum disorders activate Cav1.3 L-type calcium channels. Biol Psychiatr. 2015;77:816–822. doi: 10.1016/j.biopsych.2014.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pitt GS, Matsui M, Cao C. Voltage-gated calcium channels in non-excitable tissues. Annu Rev Physiol. 2021;83:183–203. doi: 10.1146/annurev-physiol-031620-091043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Purcell SM, Moran JL, Fromer M, Ruderfer D, Solovieff N, Roussos P, O’Dushlaine C, Chambert K, Bergen SE, Kahler A, Duncan L, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506:185–190. doi: 10.1038/nature12975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodan LH, Spillmann RC, Kurata HT, Lamothe SM, Maghera J, Jamra RA, Alkelai A, Antonarakis SE, Atallah I, Bar-Yosef O, Bilan F, et al. Phenotypic expansion of CACNA1C-associated disorders to include isolated neurological manifestations. Genet Med. 2021;23:1922–1932. doi: 10.1038/s41436-021-01232-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaw RJ, Mackay D, Pell JP, Padmanabhan S, Bailey DS, Smith DJ. The relationship between antihypertensive medications and mood disorders: analysis of linked healthcare data for 1.8 million patients. Psychol Med. 2021;51:1183–1191. doi: 10.1017/S0033291719004094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh T, Poterba T, Curtis D, Akil H, Al Eissa M, Barchas JD, Bass N, Bigdeli TB, Breen G, Bromet EJ, Buckley PF, et al. Rare coding variants in ten genes confer substantial risk for schizophrenia. Nature. 2022;604:509–516. doi: 10.1038/s41586-022-04556-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sklar P, Smoller JW, Fan J, Ferreira MA, Perlis RH, Chambert K, Nimgaonkar VL, McQueen MB, Faraone SV, Kirby A, de Bakker PI, et al. Whole-genome association study of bipolar disorder. Mol Psychiatr. 2008;13:558–569. doi: 10.1038/sj.mp.4002151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldatov NM, Bouron A, Reuter H. Different voltage-dependent inhibition by dihydropyridines of human Ca2+ channel splice variants. J Biol Chem. 1995;270:10540–10543. doi: 10.1074/jbc.270.18.10540. [DOI] [PubMed] [Google Scholar]
- Song JHT, Lowe CB, Kingsley DM. Characterization of a human-specific tandem repeat associated with bipolar disorder and schizophrenia. Am J Hum Genet. 2018;103:421–430. doi: 10.1016/j.ajhg.2018.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R, Napolitano C, Schwartz PJ, Joseph RM, Condouris K, Tager-Flusberg H, et al. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell. 2004;119:19–31. doi: 10.1016/j.cell.2004.09.011. [DOI] [PubMed] [Google Scholar]
- Striessnig J, Grabner M, Mitterdorfer J, Hering S, Sinnegger MJ, Glossmann H. Structural basis of drug binding to L Ca2+ channels. Trends Pharmacol Sci. 1998;19:108–115. doi: 10.1016/s0165-6147(98)01171-7. [DOI] [PubMed] [Google Scholar]
- Striessnig J, Pinggera A, Kaur G, Bock G, Tuluc P. L-Type Ca(2 +) Channels in Heart and Brain. Wiley Interdiscip Rev Membr Transp Signal. 2014;3:15–38. doi: 10.1002/wmts.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang ZZ, Hong X, Wang J, Soong TW. Signature combinatorial splicing profiles of rat cardiac- and smooth-muscle Cav1.2 channels with distinct biophysical properties. Cell Calcium. 2007;41:417–428. doi: 10.1016/j.ceca.2006.08.002. [DOI] [PubMed] [Google Scholar]
- Tang L, Gamal El-Din TM, Swanson TM, Pryde DC, Scheuer T, Zheng N, Catterall WA. Structural basis for inhibition of a voltage-gated Ca(2 +) channel by Ca(2 +) antagonist drugs. Nature. 2016;537:117–121. doi: 10.1038/nature19102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tao R, Cousijn H, Jaffe AE, Burnet PW, Edwards F, Eastwood SL, Shin JH, Lane TA, Walker MA, Maher BJ, Weinberger DR, et al. Expression of ZNF804A in human brain and alterations in schizophrenia, bipolar disorder, and major depressive disorder: a novel transcript fetally regulated by the psychosis risk variant rs1344706. JAMA Psychiatr. 2014;71:1112–1120. doi: 10.1001/jamapsychiatry.2014.1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiwari S, Zhang Y, Heller J, Abernethy DR, Soldatov NM. Atherosclerosis-related molecular alteration of the human CaV1.2 calcium channel alpha1C subunit. Proc Natl Acad Sci U S A. 2006;103:17024–17029. doi: 10.1073/pnas.0606539103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trubetskoy V, Pardinas AF, Qi T, Panagiotaropoulou G, Awasthi S, Bigdeli TB, Bryois J, Chen CY, Dennison CA, Hall LS, Lam M, et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature. 2022;604:502–508. doi: 10.1038/s41586-022-04434-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vahdani B, Kian AA, Esmaelizadeh A, Zenoozian S, Yousefi V, Mazloomzadeh S. Adjunctive raloxifene and isradipine improve cognitive functioning in patients with schizophrenia: a pilot study. J Clin Psychopharmacol. 2020;40:457–463. doi: 10.1097/JCP.0000000000001274. [DOI] [PubMed] [Google Scholar]
- Wainberg M, Merico D, Keller M>C, Fauman EB, Tripathy SJ. Predicting causal genes from psychiatric genome-wide association studies using high-level etiological knowledge. Mol Psychiatr. 2022;27:3095–2106. doi: 10.1038/s41380-022-01542-6. [DOI] [PubMed] [Google Scholar]
- Wang Y, Tang S, Harvey KE, Salyer AE, Li TA, Rantz EK, Lill MA, Hockerman GH. Molecular determinants of the differential modulation of Cav1.2 and Cav1.3 by nifedipine and FPL 64176. Mol Pharmacol. 2018;94:973–983. doi: 10.1124/mol.118.112441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Chen R, Cheng F, Wei Q, Ji Y, Yang H, Zhong X, Tao R, Wen Z, Sutcliffe JS, Liu C, et al. A Bayesian framework that integrates multi-omics data and gene networks predicts risk genes from schizophrenia GWAS data. Nat Neurosci. 2019;22:691–699. doi: 10.1038/s41593-019-0382-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Horigane SI, Wakamori M, Ueda S, Kawabata T, Fujii H, Kushima I, Kimura H, Ishizuka K, Nakamura Y, Iwayama Y, et al. Identification of ultra-rare disruptive variants in voltage-gated calcium channel-encoding genes in Japanese samples of schizophrenia and autism spectrum disorder. Transl Psychiatry. 2022;12:84. doi: 10.1038/s41398-022-01851-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wankerl K, Weise D, Gentner R, Rumpf JJ, Classen J. L-type voltage-gated Ca2+ channels: a single molecular switch for long-term potentiation/long-term depression-like plasticity and activity-dependent metaplasticity in humans. J Neurosci. 2010;30:6197–6204. doi: 10.1523/JNEUROSCI.4673-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, Yan Z, Li Z, Yan C, Lu S, Dong M, Yan N. Structure of the voltage-gated calcium channel Cav1.1 complex. Science. 2015;350:aad2395. doi: 10.1126/science.aad2395. [DOI] [PubMed] [Google Scholar]
- Xiao X, Chang H, Li M. Molecular mechanisms underlying noncoding risk variations in psychiatric genetic studies. Mol Psychiatr. 2017;22:497–511. doi: 10.1038/mp.2016.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu W, Lipscombe D. Neuronal CaV1.3α1 L-type channels activate at relatively hyperpolarized membrane potentials and are incompletely inactivated by dihydropyridines. J Neurosci. 2001;21:5944–5951. doi: 10.1523/JNEUROSCI.21-16-05944.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshimizu T, Pan JQ, Mungenast AE, Madison JM, Su S, Ketterman J, Ongur D, McPhie D, Cohen B, Perlis R, Tsai LH. Functional implications of a psychiatric risk variant within CACNA1C in induced human neurons. Mol Psychiatr. 2015;20:162–169. doi: 10.1038/mp.2014.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov. 2016;15:19–34. doi: 10.1038/nrd.2015.5. [DOI] [PubMed] [Google Scholar]
- 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;67:821–870. doi: 10.1124/pr.114.009654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang CY, Xiao X, Zhang Z, Hu Z, Li M. An alternative splicing hypothesis for neuropathology of schizophrenia: evidence from studies on historical candidate genes and multi-omics data. Mol Psychiatr. 2022;27:95–112. doi: 10.1038/s41380-021-01037-w. [DOI] [PubMed] [Google Scholar]
- Zuhlke RD, Bouron A, Soldatov NM, Reuter H. Ca2+ channel sensitivity towards the blocker isradipine is affected by alternative splicing of the human alpha1C subunit gene. FEBS Lett. 1998;427:220–224. doi: 10.1016/s0014-5793(98)00425-6. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the article.

