2,631
Views
2
CrossRef citations to date
0
Altmetric
Review

L-type calcium channels and neuropsychiatric diseases: Insights into genetic risk variant-associated genomic regulation and impact on brain development

, & ORCID Icon
Article: 2176984 | Received 07 Oct 2022, Accepted 01 Feb 2023, Published online: 19 Feb 2023

ABSTRACT

Recent human genetic studies have linked a variety of genetic variants in the CACNA1C and CACNA1D genes to neuropsychiatric and neurodevelopmental disorders. This is not surprising given the work from multiple laboratories using cell and animal models that have established that Cav1.2 and Cav1.3 L-type calcium channels (LTCCs), encoded by CACNA1C and CACNA1D, respectively, play a key role in various neuronal processes that are essential for normal brain development, connectivity, and experience-dependent plasticity. Of the multiple genetic aberrations reported, genome-wide association studies (GWASs) have identified multiple single nucleotide polymorphisms (SNPs) in CACNA1C and CACNA1D that are present within introns, in accordance with the growing body of literature establishing that large numbers of SNPs associated with complex diseases, including neuropsychiatric disorders, are present within non-coding regions. How these intronic SNPs affect gene expression has remained a question. Here, we review recent studies that are beginning to shed light on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic and chromatin levels. We additionally review recent studies that are uncovering how altered calcium signaling through LTCCs impact some of the neuronal developmental processes, such as neurogenesis, neuron migration, and neuron differentiation. Together, the described changes in genomic regulation and disruptions in neurodevelopment provide possible mechanisms by which genetic variants of LTCC genes contribute to neuropsychiatric and neurodevelopmental disorders.

Overview of CACNA1C AND CACNA1D genetic risk variants

L-type calcium channels (LTCCs) are a subgroup of voltage-gated calcium channels that include four members: Cav1.1–1.4. The most prominent forms in the brain are Cav1.2 and Cav1.3, encoded by the genes CACNA1C and CACNA1D, respectively [Citation1]. Recent genetic studies have implicated CACNA1C and CACNA1D in neurodevelopmental and neuropsychiatric diseases (with neurodevelopmental origins [Citation2–5]). The first link of LTCCs to human brain disease was the discovery of the Cav1.2 single-point mutation G406R in Timothy Syndrome, a multi-organ disorder that includes cardiac and neurological autism spectrum disorder (ASD)-like behavioral symptoms [Citation6]. Soon after, G402S was identified [Citation7], followed by I1166T [Citation8], both manifesting ASD-like symptoms. More recently, multiple additional Cav1.2 mutations have been identified in patients with a variety of symptoms, including new mutations linked to both cardiac and ASD symptoms, while others not causing ASD [Citation9]. Studies have begun to examine the impact of the various mutations on Cav1.2 channel properties to gain deeper knowledge on possible mechanisms that result in the cardiac versus brain symptoms [Citation10]. We direct readers to excellent reviews by Marcantoni et al. 2020 [Citation9] and Herold et al. 2023 [Citation11] on this topic. In the neurodevelopmental section below, we focus on Cav1.2 mutations that have been linked to ASD phenotypes in patients. Similar to Cav1.2, missense coding genetic variants in CACNA1D (Cav1.3) have been linked to ASD and intellectual disability [Citation12]. These disease-linked mutations have direct effects on channel gating and are believed to underlie the observed ASD-associated phenotypes. We direct readers to the excellent reviews by Ortner et al. 2020 [Citation12] and Ortner 2023 [Citation13] on Cav1.3 mutations and their impact on channel properties.

In addition to the coding variants, non-coding single nucleotide polymorphisms (SNPs) () in CACNA1C have been found in Genome-Wide Association Studies (GWASs) () to be associated with bipolar disorder (BD) [Citation14–17], schizophrenia (SCZ) [Citation18], major depressive disorder (MDD) [Citation19], ASD [Citation20], and attention deficit hyperactivity disorder (ADHD) [Citation21]. Similarly, SNPs in CACNA1D have been linked to BD, SCZ, ASD, and intellectual disability (ID) [Citation22–24]. The majority of these SNPs are present in intronic regions, particularly for CACNA1C. This is in accordance with the growing body of literature establishing that SNPs associated with complex diseases, such as neuropsychiatric disorders are present within non-coding genomic regions [Citation25]. The mechanisms by which these SNPs impact gene expression, and in turn channel function, have remained a question. Since many neuropsychiatric disease-associated SNPs are located in these non-coding regions of the genome, research focus has shifted to examining the influence of intronic SNPs in the context of broader genomic regulation [Citation26–28]. This includes studies on how disease-linked SNPs present within cis-regulatory elements that include enhancers and promoters (), impact chromatin at a three-dimensional (3D) level and subsequently impact gene expression (as discussed in the next section).

Box 1. Definitions of terms

In this review, we will discuss new literature on how neuropsychiatric-linked non-coding genetic variants can impact gene expression via regulation at the genomic level, with a focus on CACNA1C, which has been studied more over the last several years compared to CACNA1D. We additionally review recent findings on how aberrant Cav1.2 and Cav1.3 channel expression or function can impact brain development as studied in both in vitro and in vivo models of brain development. Readers are directed to previous reviews that have covered CACNA1C and CACNA1D genetic variants, animal studies, molecular signaling mechanisms of these channels, and their contribution to neuropsychiatric-related cognitive domains [Citation9,Citation22,Citation23,Citation30–34].

Intronic non-coding SNPs and genomic regulation

The development of new technologies, such as chromosome conformation capture [Citation35,Citation36] (), which measures physical interactions in the genome to probe the 3D mammalian genome, has greatly advanced our understanding of the structural and functional dynamics of the genome and how genes are transcribed [Citation37,Citation38]. In eukaryotic cells, DNA is in a compact form, with 146–147 base pairs of DNA wrapped around an octamer of histones, making up nucleosomes that are further condensed into chromatin, which is what makes up a chromosome [Citation39–41] (). It is now clear that although genetic information is encoded in DNA’s linear sequences, transcriptional regulation is dictated by the 3D hierarchical organization of chromatin bringing together different DNA domains across multiple kilobases in close proximity to each other, to activate gene expression (; see Fujita et al., 2022 [Citation42] for review).

Figure 1. Gene expression regulatory processes at the level of chromatin that can be disrupted by disease-associated SNPs as observed in LTCC genes CACNA1C and CACNA1D. Gene expression can be regulated at the level of (1) DNA methylation, (2) histone modifications, (3) miRNA, and (4) splicing. Additionally, structural changes at the genomic level, such as chromatin looping which brings regulatory DNA elements such as enhancers and promoters in close proximity, are involved in activation transcription. The protein CTCF (CCCTC DNA-binding factor) plays a key role in changes in chromatin architecture.

Figure 1. Gene expression regulatory processes at the level of chromatin that can be disrupted by disease-associated SNPs as observed in LTCC genes CACNA1C and CACNA1D. Gene expression can be regulated at the level of (1) DNA methylation, (2) histone modifications, (3) miRNA, and (4) splicing. Additionally, structural changes at the genomic level, such as chromatin looping which brings regulatory DNA elements such as enhancers and promoters in close proximity, are involved in activation transcription. The protein CTCF (CCCTC DNA-binding factor) plays a key role in changes in chromatin architecture.

With the majority of the genome consisting of non-coding DNA and the complexity of 3D chromatin structure, studies have shifted to identifying transcriptional regulatory regions at the gene level. This has allowed looking at how 3D interactions are disrupted by non-coding genetic variants in complex diseases, including neuropsychiatric diseases [Citation43]. Recent large-scale initiatives such as the ENCODE (ENCyclopedia of DNA Elements) consortium have found that non-coding intronic regions are rich in regulatory elements including enhancers, repressors, silencers, and insulators () that can influence gene expression. These intronic elements interact with cis-regulatory elements, such as promoters (), and transcription factor binding sites classically present at the 5’ end of genes, through long-range structural chromatin modifications () [Citation38,Citation44]. Many disease-linked SNPs are present within these non-coding regions of the DNA that have the potential to disrupt normal transcriptional mechanisms [Citation45].

Other mechanisms that occur either before or in tandem with the structural chromatin changes include epigenetic changes such as DNA methylation and histone modifications (). At the level of the RNA transcript, microRNAs and alternative splicing further impact mRNA levels or types of transcripts generated. Non-coding SNPs can dysregulate any of the mechanisms described above, in turn impacting gene expression. Below we review some new literature characterizing the genomic landscape of the CACNA1C gene and the interaction of non-coding SNPs with genomic elements to influence gene expression. Such knowledge is proving to be critical for understanding the genetics of complex disorders, such as genetic findings from GWASs of neuropsychiatric disorders [Citation46–48].

Chromatin looping

One of the structural changes that promotes transcriptional activation is the looping of DNA, referred to as chromatin looping, which regulates transcription by changing the proximity of different regulatory elements [Citation49]. For example, a chromatin loop can bring an enhancer that is thousands of base pairs away from the promoter of the gene in close proximity to increase gene expression (). Thus, intronic SNPs within these regulatory regions can disrupt chromatin looping by affecting the accessibility of DNA-binding factors necessary for chromatin looping [Citation50–52]. Disruptions in chromatin looping have been implicated in neurological disorders, such as schizophrenia, Alzheimer’s disease, and addiction, as reviewed by Behrends and Engmann [Citation51].

Specific to mouse Cacna1c, a few studies have highlighted the importance of chromatin looping in transcriptional activation and the impact of intronic SNPs. A demonstration for the importance of looping has come from the study of the zinc finger protein CCCTC DNA-binding factor (CTCF), a well-defined regulator of chromatin architecture. Mice with a knockout of CTCF in cardiomyocytes have a loss of almost all global chromatin loops, which in turn leads to decreased Cacna1c expression in the heart [Citation53]. Though only demonstrated in the heart, this regulation of Cacna1c expression through chromatin loops could occur similarly in the brain.

Important to the genomic regulation of human CACNA1C, the third intron of CACNA1C contains SNPs linked to multiple neuropsychiatric diseases [Citation21]. One of the first observations that disease-associated SNPs could impact gene regulation came from the work of Roussos et al. (2014) showing that multiple SCZ-associated SNPs in the third intron are in an enhancer region that interacts with the proximal gene promoter [Citation54]. Using chromosome conformation capture (; 3C-seq and 4C-seq;), which characterizes the spatial organization of chromatin and finds interactions between genomic loci [Citation55], they identified that a region in intron 3 interacted with the CACNA1C promoter through chromatin looping [Citation54]. The intronic region contained the strongest SCZ risk SNP (rs1006737) and majority of the CACNA1C SNPs that are in high linkage disequilibrium (). These SNPs disrupted chromatin looping, decreasing transcriptional activity and reducing CACNA1C mRNA levels. This demonstrated a mechanism by which intronic SNPs could disrupt chromatin looping and gene expression [Citation54]. They observed this in human post-mortem SCZ brains, human induced pluripotent cells, HEK, and SKN-SH cell lines [Citation54]. Additional regions have been found in the third intron of CACNA1C with enhancer activity, which could also regulate transcriptional activity of the gene [Citation56]. Eckart et al. (2016) have similarly demonstrated with chromatin conformation capture that there is an interaction of a SCZ-associated region in intron 3, with the CACNA1C promoter and other potential regulatory regions that influence gene expression [Citation57]. This region within intron 3 contains 16 SNPs that are in linkage disequilibrium with the previously mentioned SCZ risk SNP rs1006737 that could also disrupt chromatin looping. These studies demonstrate that disruption of chromatin looping required for normal gene expression could be one way in which intronic SNPs lead to changes in CACNA1C gene expression that could contribute to neuropsychiatric disease.

Epigenetics

Epigenetic modifications regulate the genome and have many downstream effects through altering gene expression without changing the DNA sequence [Citation58,Citation59]. These modifications include classical chromatin altering mechanisms, such as DNA methylation and post-translational modifications of histones, as well as non-classical mechanisms, such as regulation by non-coding microRNAs (). All of these modes of genome regulation have been associated with neuropsychiatric disorders, including MDD, ASD, Fragile X syndrome, Rett syndrome, BD, and SCZ [Citation60].

A. DNA methylation

DNA methylation is the addition of a methyl group to the cytosine of a CG dinucleotide (termed CpG site) to form 5-methylcytosine [Citation61], and is one of the most-studied epigenetic mechanisms. Alterations of DNA methylation levels can play a direct role in regulating gene expression [Citation62,Citation63], alternative splicing [Citation64–66], and alternative promoter () usage [Citation67,Citation68]. DNA methylation is a dynamic process within cells that is regulated by DNA methyltransferases that are cytosine methylases belonging to a conserved family of proteins [Citation69]. DNMT1 is the maintenance methyltransferase, responsible for copying DNA methylation from the old DNA strand to the new one during cell division, which in the brain occurs during development prior to neural stem cells becoming mature neurons, while DNMT3A and DNMT3B are responsible for de novo activity-dependent DNA methylation [Citation70–72]. A study in mouse embryonic cells overexpressing DNMT1 showed lower levels of Cacna1c mRNA levels [Citation73], demonstrating that altered level of DNA methylation can influence transcription of the Cacna1c gene.

SNPs can occur either within or nearby CpG sites, disrupting normal DNA methylation, either directly or indirectly via altering genome-level interactions required for transcription [Citation74], as described in previous sections. Differential DNA methylation of the CACNA1C gene and the interaction between methylation and SNPs has been observed in BD [Citation75], a disorder with well-characterized CACNA1C SNPs [Citation14–17]. When analyzing 169 CpG sites spread across five CpG islands (CGIs; ) in the CACNA1C gene using DNA isolated from the blood of BD patients, Starnawska et al. (2016) found that one of the five CGIs studied demonstrated intermediate levels of methylation compared to that of healthy controls [Citation75]. Within this one CGI, five of the six CpG sites showed hypermethylation in patient samples. The study additionally examined the interaction of BD-associated SNPs with the observed hypermethylation, and found that SNP rs2238056 in intron 3 had the strongest methylation quantitative trait locus (), pointing to a mechanism of how SNPs can interact with DNA methylation status to influence CACNA1C gene expression [Citation75].

Differential methylation of genes including CACNA1C have also been found in patients with psychosis in an Epigenome-Wide Association Study (EWAS; ) [Citation76]. The CpG at the intronic position cg01833890 in CACNA1C was found to be differentially methylated in patients exhibiting psychosis [Citation76]. Similarly, the rs1990322 locus in CACNA1C at cg24393317 was significantly associated with PTSD [Citation77] and variable methylation was found between depression-discordant monozygotic twins at position cg10031793 in CACNA1C [Citation78]. A methylation quantitative trait locus (significant SNP-CpG methylation association), rs2283291, was also found in CACNA1C using DNA isolated from postmortem prefrontal cortex tissue from patients with SCZ [Citation79] and was associated with SCZ in a study of Chinese men [Citation80]. All of these studies, except for the ones looking in postmortem tissue, looked at methylation in DNA isolated from patient blood. Even though it is difficult to know if methylation status in blood cells parallels brain methylation status, these studies suggest that SNPs can influence methylation status of the CACNA1C gene, which could alter gene expression levels.

B. Histone modifications

Post-translational modifications of histones () can affect how the DNA is able to wrap around the histone, changing accessibility to DNA and thus influencing gene transcription [Citation81]. Histone modifications promote altered levels of chromatin condensation, resulting in either “open” (referred to as euchromatin) or “closed” (referred to as heterochromatin) states of chromatin, that increase or decrease transcription, respectively [Citation82,Citation83].

Histone acetylation and methylation are the most studied post-translational modifications. H3 mono-methylation of lysine 4 (H3K4me1) and H3 acetylation of lysine 27 (H3K27ac) are associated with the “open” chromatin state and promote transcription [Citation84,Citation85]. Supporting regulation of CACNA1C by histone modifications is the presence of H3K4me1 in an enhancer region within intron 3 of CACNA1C [Citation86]. In BD patients, seven disease-associated SNPs are present within this intron 3 enhancer [Citation44] that could modify histone binding and alter CACNA1C mRNA expression, as has been reported for schizophrenia-associated SNPs also present within this region [Citation54].

The importance of this CACNA1C intron 3 enhancer and histone modification is underscored by the observation that one of the SCZ-associated risk SNP in this region interacts via long-range chromatin looping with the CACNA1C proximal promoter [Citation54,Citation57]. The functional significance of this interaction comes from the finding that the promoter region harbors a binding site for the epigenetic regulator, EZH2 (Enhancer of zeste homolog 2) [Citation87]. EZH2 is a histone-lysine N-methyltransferase enzyme that participates in histone methylation by addition of methyl groups to histone H3 at lysine 27, resulting in transcriptional repression. Consistent with EZH2ʹs role as a repressor, in vitro studies in the SH-SY5Y cell line have found that EZH2 represses reporter expression of a construct containing the CACNA1C promoter. The importance of EZH2 and thus histone modifications to CACNA1C in SCZ and BD is further highlighted by the observation that expression analysis from the anterior cingulate cortex region of SCZ and BD patient brains showed highly upregulated EZH2 that correlated with downregulated expression of CACNA1C [Citation88]. Thus, EZH2, a histone modifying enzyme, through its actions at the CACNA1C promotor, and/or through disrupting interactions with enhancer-containing SNPs, could impact CACNA1C expression.

C. microRNA

MicroRNAs (miRNAs) are a type of short non-coding RNAs, 21–26 nucleotides long, that can regulate gene expression [Citation89]. They do so through binding to complementary mRNA, often at the 3’ untranslated region (UTR) and decrease gene expression by repressing translation or degrading mRNA [Citation89,Citation90]. Converging animal and human studies have identified a key role of microRNAs in neuropsychiatric disorders [Citation91,Citation92].

CACNA1C is a known target of a miRNA, microRNA137 (MIR137) [Citation93,Citation94], that has been linked to SCZ [Citation95]. In HEK-293T cells, transfection of miR-137 results in decreased CACNA1C expression [Citation94,Citation96]. It turns out that EZH2, described above, is also a validated target of MIR137 [Citation97,Citation98], thus it is possible that the miR-137 could also regulate CACNA1C indirectly via changes in EZH2 expression. Functional studies have demonstrated that increasing expression of miR-137 in adult neural stem cells isolated from mice in vitro increases cell proliferation, whereas decreasing miR-137 reduces proliferation. Similarly, increasing miR-137 expression in the adult mouse hippocampus in vivo led to increased adult hippocampal neurogenesis [Citation99], a mechanism regulated by cacna1c [Citation100] and altered in neuropsychiatric disorders [Citation101–103]. In addition, in mouse hippocampal neurons, miR-137 regulates synapse formation, maturation, and transmission [Citation104]. Therefore, aberrations in MIR137 in neuropsychiatric conditions could be due to altered expression of CACNA1C and related genes that impacts brain cellular and synaptic mechanisms in the hippocampus. Additionally, another miRNA, miR-4300, which also targets CACNA1C, was found to be enriched in copy number variable areas, which is a section of the genome that has a variable number of copies between individuals, in a treatment-resistant SCZ patient [Citation105].

D. Splicing

Recent studies have underscored alternative splicing as a key molecular process that vastly expands the proteome [Citation106]. Interestingly, dysregulation of alternative splicing has been linked to neuropsychiatric [Citation107–109] and neurodevelopmental disorders [Citation110,Citation111]. Not surprisingly, alternative splicing is regulated by multiple epigenetic influences, including DNA methylation [Citation64,Citation65,Citation112] and disease-linked SNPs [Citation113,Citation114], showing that genetic variants could impact normal splicing and contribute to neuropsychiatric diseases.

Recent developments in long-read sequencing have allowed for the characterization of the deep complexity of human CACNA1C splicing, identifying 38 novel exons and 241 novel transcripts [Citation115]. There are variations in splice isoforms across different brain regions, which could help differentiate functional differences in the variants [Citation115]. Because many SNPs are in non-coding regions of CACNA1C [Citation25], it reasons that SNPs occurring at splice sites within mRNAs could impact splicing and thus could affect transcriptional and translational regulation of the gene.

One of the clearest examples of the effect of splicing occurs in TS in the CACNA1C gene, where the G406R mutation in the alternatively spliced exon 8A of Cacna1c leads to aberrant splicing of exons 8 and 8A, which is regulated during cortical development [Citation116]. The G406R mutation prevents a normal developmental switch of exon utilization from exon 8A to exon 8, which leads to Timothy Syndrome 1 and Timothy Syndrome 2, respectively. Below we describe the impact of TS mutations on brain development.

Even though much less studied than CACNA1C, changes in the splicing of Cacna1d have been reported in a zebrafish model of SCZ [Citation117]. In larval zebrafish, heterozygotes with the splice mutation sa17298 in Cacna1d results in 50% reduction of splice variants 201 and 202 [Citation117]. This reduction in splice variants led to “psychosis-like” behaviors, including startle responses to dark flashes and locomotor activity under constant light that could be reversed with antipsychotics, such as risperidone and haloperidol [Citation117]. This finding demonstrated a role for Cacna1d in SCZ-relevant behaviors and a potential schizophrenia-like model in zebrafish for studying the role of Cacna1d splicing.

L-type calcium channels and brain development

Major steps in cortical development

It is critical that proper genetic programs are recruited for normal brain development. The mammalian central nervous system is comprised of highly interconnected brain regions that carry out a variety of higher cognitive, emotional, and sensorimotor functions that are disrupted in neuropsychiatric and neurodevelopmental disorders [Citation118]. In particular, the cerebral cortex, a widespread sheet of neural tissue at the outermost region of the cerebrum, plays a key role in the execution of complex behaviors [Citation118]. The cerebral cortex consists of six layers, which contain groups of neurons that are genetically, morphologically, neurochemically, and electrophysiologically distinct [Citation118]. Furthermore, each layer makes connections to and from distinct cortical and subcortical regions that allow communication between different brain regions. Given the complexity of the organization and function of the cerebral cortex, it is critical that the six-layer structure, as well as its connections, are properly assembled during development. Establishing the cytoarchitecture and neural circuits of the cerebral cortex, as well as other brain regions, requires numerous developmental processes that need to be temporally and spatially regulated (). The process can be grossly divided into four steps: (1) generation of neurons and glia (neurogenesis and gliogenesis), (2) specification of identity and spatial location of neurons and glia (differentiation and migration), (3) forming connections with other neurons (synaptogenesis, axonogenesis, myelination), (4) experience-dependent maturation of neural circuits.

Figure 2. Major steps in mammalian cortical development. (a) Coronal diagram of the embryonic mouse brain at embryonic day 12 (E12). (b) Zoom in of dotted box in (a), showing major steps of neurogenesis and gliogenesis. LGE=lateral ganglionic eminence; MGE=medial ganglionic eminence; NP=neural progenitors; RGC=radial glial cells; SVZ=subventricular zone; VZ=ventricular zone; IPC=intermediate progenitor cells; L=layer.

Figure 2. Major steps in mammalian cortical development. (a) Coronal diagram of the embryonic mouse brain at embryonic day 12 (E12). (b) Zoom in of dotted box in (a), showing major steps of neurogenesis and gliogenesis. LGE=lateral ganglionic eminence; MGE=medial ganglionic eminence; NP=neural progenitors; RGC=radial glial cells; SVZ=subventricular zone; VZ=ventricular zone; IPC=intermediate progenitor cells; L=layer.

Genetic mutations that occur in genes that are critical for cortical development can lead to aberrant development of the cerebral cortex layering, circuit wiring, and ultimately, function. These aberrations lead to behavioral impairments and increased susceptibility to neuropsychiatric disorders including SCZ and ASD [Citation15,Citation119–121]. Given the link between LTCCs and neuropsychiatric disorders [Citation2,Citation22,Citation23,Citation34] and the impact of CACNA1C and CACNA1D mutations on channel function (as in Timothy syndrome [Citation6,Citation8,Citation12] and ASD [Citation24], respectively) and of CACAN1C SNPs on gene expression, it is critical to understand the contribution of LTCCs in cerebral cortex development. Here, we review how perturbations in LTCC expression and/or function impacts different developmental processes.

Neurogenesis and gliogenesis

The initial step of cortical development is generating the proper number of neurons and glia (astrocytes, oligodendrocytes, and microglia). Based on their function, neurons in the cerebral cortex can be divided into excitatory or inhibitory neurons. Excitatory neurons in the cerebral cortex originate from a layer of neural stem cells above the lateral ventricles known as the ventricular zone (VZ) (), while inhibitory neurons originate from a group of stem cells below the lateral ventricles known as the ganglionic eminence (GE) (). Before embryonic day (E) 10, neural stem cells in the VZ undergo mitosis (i.e. proliferate) to generate a sufficient number of neural progenitors. Thereafter, neural progenitors take an elongated morphology (radial glial cells; RGC, ) to form a radial scaffold that guides the newborn neurons to their proper location. RGCs not only help the migration of neurons, but as a neural stem cell, they give rise to cortical excitatory pyramidal neurons to build a layered cytoarchitecture. From E11 through E13, one RGC will divide into another RGC, as well as an immature neuron that migrate to the deep layers of the cerebral cortex (layers 5 and 6). From E13 through E16, one RGC will divide into another RGC, as well as an intermediate progenitor. These intermediate progenitors will divide and differentiate into neurons that migrate to the upper layers of the cerebral cortex (layers 1–4). After neurogenesis is completed by E16, starting at E17, the RGCs become astrocytes, as well as give rise to oligodendrocyte progenitor cells (OPCs) ().

Mice overexpressing the TS G406R mutation in neural progenitor cells (using Foxg1Cre) do not show differences in the total number of neurons in the somatosensory cortex at postnatal day 0, suggesting that neurogenesis is not affected by aberrant calcium signaling [Citation122]. In the same study, TS patient-derived neural progenitor cells did not show differences in proliferation in vitro [Citation122]. Furthermore, when TS G406R mutant Cav1.2 channels (Cav1.2-G406R) or calcium-impermeable Cav1.2 channels (Cav1.2-G406R-4EQ) were overexpressed into the neural progenitors of the VZ via in utero electroporation, both manipulations did not alter the number of RGCs or intermediate progenitors. TS patient-derived in vitro cerebral organoids that resemble the GE, from which inhibitory interneurons originate, show no differences in expression of progenitor markers. Together, these studies suggest that Cav1.2 gain-of-function does not lead to changes in neurogenesis of both excitatory and inhibitory neurons of the cerebral cortex. Similar to neurogenesis, mice carrying a heterozygous mutation for Cav1.2-G406R do not show differences in the number of oligodendrocytes as well as the proliferation of oligodendrocyte progenitors [Citation123]. However, whether gliogenesis of astrocytes or microglia are affected by the Cav1.2-G406R gain-of-function mutation remains to be studied.

While the impact of loss of Cacna1c (Cav1.2) on neurogenesis is unknown, given that the expression of Cacna1c is weak in the VZ throughout mouse embryonic development (see Panagiotakos et al., 2019 [Citation116] for E11, 14, 16, and postnatal day (P)1 and Horigane et al., 2020 [Citation124] for E17) in which the neural progenitors reside, it is likely that loss of Cacna1c (Cav1.2) would not impact neurogenesis. Interestingly, male mice and rats with heterozygous loss of Cacna1c or male mice with Cacna1c loss in excitatory cells show reduced adult neurogenesis in the hippocampus [Citation31,Citation100,Citation125,Citation126]. In contrast, in female rats with heterozygous loss of Cacna1c had no effect on adult hippocampal neurogenesis [Citation127]. However, the impact of heterozygous loss of Cacna1c on developmental neurogenesis and gliogenesis in the cerebral cortex remains unknown. Cacna1d also shows low expression in the VZ at E17 [Citation124], but as there is limited evidence in the cell-type specific expression pattern of Cacna1d across developmental stages in the embryonic cortex (see Schlick, Flucher and Obermair, 2010 [Citation128] for whole cortex mRNA expression), the impact of heterozygous loss of Cacna1d on cerebral cortical neurogenesis and gliogenesis is difficult to speculate. Nonetheless, similar to mice with heterozygous loss of Cacna1c, mice with heterozygous loss of Cacna1d also have reduced adult neurogenesis in the hippocampus [Citation129,Citation130].

Neuronal migration

As neurons are born from neural progenitors, they migrate to their final position in the embryonic cortex and take on a distinct identity. Cortical excitatory and inhibitory neurons use different modes of migration to reach their final location in the developing cortex. Excitatory pyramidal neurons migrate radially from the VZ to either the upper layer or deep layer of the developing cortex (referred to as radial migration; ). Inhibitory interneurons that originate from the GE tangentially migrate to reach the embryonic cortex to find their laminar position (referred to as tangential migration [Citation118];; ). Both routes of migration require regulation of cell adhesion molecules and actin cytoskeleton. While many mechanisms control transcription factors and cytoskeletal activity during differentiation and migration, calcium signaling is a key component. In particular, calcium activity, measured by a calcium indicator, is increased in migrating neurons when treated with LTCC activators, such as FPL64176 [Citation131,Citation132] or decreased when treated with LTCC blocker nimodipine [Citation131], suggesting LTCC-dependent calcium signaling may play a role in neuronal migration.

Overexpression of wild-type Cav1.2 (Cav1.2-WT) and reporter construct (EGFP) in upper layer excitatory neurons being born (using in utero electroporation at E15) leads to more EGFP-expressing neurons in deep layers compared to expression of an empty vector [Citation131]. Furthermore, overexpression of Cav1.2-G406R channels lead to more EGFP-expressing neurons in deep layers compared to the control vector, but also significantly more than when Cav1.2-WT was overexpressed [Citation131]. Similarly, when a Cav1.2 with a different mutation found in TS-like patients (Cav1.2-I1166T) was overexpressed (with a GFP-reporter), in upper layer excitatory neurons being born, more GFP-expressing neurons were found in the deep layers compared to control [Citation133].

Radial migration deficits due to aberrant calcium signaling (via Cav1.2-G406R or Cav1.2-I1166T) were reversed by decreasing calcium permeability of the mutated Cav1.2 channel (Cav1.2-G406R-4EQ [Citation131]; Cav1.2-I1166T-L745P [Citation133]). Furthermore, a TS-like Cav1.2-I1166T mutation-induced migration deficit was reversed when Cav1.2-α1 and Cav1.2-β subunits (via W440A mutation) or calmodulin binding (via I1624A mutation) were blocked, but not when calcineurin binding (via A1929P mutation) was inhibited [Citation133]. Interestingly, the Cav1.2-G406R-induced radial migration deficit was observed when the channels were overexpressed prenatally and not when they were expressed postnatally (P1-16) [Citation131], suggesting that radial migration deficits are due to excess embryonic calcium influx in migrating neurons. Also, radial migration deficits were not seen when Cav1.2-G406R was expressed prenatally and blocked early postnatally (via Tet-ON TRE/rtTA system [Citation131], ), suggesting that once calcium influx is normalized, neurons in the wrong laminal location can continue to migrate to their proper location.

While the role of Cav1.2 LTCCs in excitatory cortical neurons are well studied, there are limited studies that indicate the role of Cav1.2 in cortical interneuron migration. In vitro studies using forebrain cerebral assembloids () derived from human induced pluripotent stem cells (iPSC, ) from TS patients show shorter saltation (each movement of migration) length and increased saltation frequency of inhibitory interneurons [Citation134,Citation135], suggesting inefficient migration of interneurons. Abnormal saltation length (but not frequency) of TS patient interneurons was rescued when treated with an LTCC blocker, nimodipine, suggesting that change in saltation length is due to excess calcium influx [Citation134,Citation135]. TS patient interneurons showed increased expression of GABA receptor subunits (α1, α4, and γ1) and treatment with the GABA antagonist picrotoxin decreased saltation frequency, implicating that saltation length and frequency are differentially regulated. In mice harboring the Cav1.2-G406R mutation (TS2-neo), more inhibitory interneurons migrating to the cortex were observed at E13.5, but the excess migration was not specific to a particular layer [Citation124]. The contrasting results between the two in vitro studies [Citation134,Citation135] and in vivo study [Citation124] may be due to many factors. One possibility is the difference in developmental expression of exon 8 containing isoforms between the TS2-neo mice [Citation136] and patient-derived neural progenitors. During normal cortical development exon 8A is the predominant isoform until E14, and exon 8 increases in expression significantly by birth [Citation116]. In TS patient-derived neural progenitor cells, exon 8A is significantly higher than exon 8 compared to control patient-derived neural progenitor cells [Citation116]. The TS2-neo mice has an inversed neomycin cassette in exon 8A which leads to reduced expression of both exon 8 and 8A [Citation124,Citation136], whereas the patient-derived assembloids bear the Cav1.2-G406R mutation in exon 8A, which leads to aberrant increase in expression of exon 8A. It is also possible that given the in vitro studies and the in vivo study measured different parameters (migration speed versus number of neurons) at different timescales (live imaging versus one time point), differences in isoform expression may not be the only contributing factor to the inconsistent results.

The aforementioned in vivo studies were conducted in the mouse somatosensory cortex, but not much is known about the developmental role of Cacna1c or Cacna1d in other cortical regions. One study showed that Cacna1c is expressed in the embryonic mouse medial prefrontal cortex (mPFC), and systemic treatment of LTCC blocker nifedipine from E13-16 reduced the number of interneurons by half (though not statistically significant compared to vehicle treated) in the embryonic mPFC [Citation137]. However, as most of the studies on Cacna1c and neuronal migration are done in the context of Cacna1c gain-of-function mutations, it is unclear how loss of Cacna1c or Cacna1d impacts neuronal migration within the somatosensory cortex as well as other brain regions.

Neuronal differentiation

As neurons migrate through the developing cortex, they acquire a number of properties that collectively comprise their fate. These properties include their final laminar location, connectivity with other brain regions, and electrical activity. The acquisition of neuronal subtype and laminar location is partly regulated by genetic programs that are specialized for each cell type [Citation138–141]. For example, two types of neurons in layer V are both born simultaneously, but their axon projection targets are different. Neurons that project to the contralateral hemisphere via the corpus callosum (callosal projection neurons, CPNs) require the expression of SATB2, whereas neurons that project to subcortical structures and spinal cord (subcerebral projection neurons, SCPNs) require the expression of FEZF2 and CTIP2 [Citation142–148].

Cortical cultures differentiated from TS patient-derived iPSCs showed significantly more neurons that express upper layer markers and significantly less neurons that express deep layer markers [Citation122]. In particular, there were significantly less SATB2-expressing neurons and more CTIP2-expressing neurons in the upper layer neurons [Citation122]. These in vitro results were recapitulated in mice overexpressing the Cav1.2-G406R mutation in neural progenitor cells (using Foxg1Cre), such that fewer SATB2+ cells were found in deep layers (V and VI) [Citation122]. In agreement with Pasca et al. (2011) [Citation122], another study showed that overexpressing Cav1.2-G406R channels in neurons that are being born at E13 (deep layer neurons) increased the number of CTIP2-expressing neurons and decreased the number of SATB2-expressing neurons [Citation116]. There were no changes in the proportion of CTIP2- and SATB2-expressing neurons when a calcium impermeable Cav1.2-G406R-4EQ channel was overexpressed, suggesting that the higher influx of calcium through mutant Cav1.2 channels contributes to the change in relative abundance of early-born neuron subtypes. Conversely, knocking out Cacna1c (Cre-expressing plasmid electroporated into Cacna1c floxed mice) at E12 decreased the number of CTIP2-expressing neurons and increased the number of SATB2-expressing neurons [Citation116]. Together, this suggests that Cav1.2 LTCCs play a critical role in the differentiation of neural progenitors to CTIP2- and SATB2-expressing cortical neurons. While the role of Cav1.2 LTCCs in neuronal differentiation was primarily studied in the developing cerebral cortex, results from the Pasca et al. (2011) in vitro studies suggest other neuronal subtypes may be altered in TS patients [Citation122]. In TS patient-derived neurons, there was a significant increase in the number of tyrosine hydroxylase (TH)-expressing neurons [Citation122]. This increase in catecholamine synthesizing enzyme was associated with increased catecholamine synthesis in TS patient-derived neurons [Citation122]. However, whether TH expression or catecholaminergic function is altered in mice overexpressing the Cav1.2-G406R mutation in the cerebral cortex or mesencephalic dopaminergic system is unknown. Furthermore, the extent of the impact of LTCCs on neuronal subtype specification across different brain regions remains to be studied.

Synaptogenesis, axonogenesis, and myelination

Differentiated neurons, as part of their identity, will make unique local and long-range connections through forming synapses with incoming neuronal inputs (synaptogenesis), axons branching out to different brain regions (axonogenesis) and supporting communication via myelination of axons. In mice harboring a heterozygous Cav1.2-G406R mutation, layer 2/3 excitatory neurons in the frontal cortex show a significant decrease in total basal dendritic length and in the number of dendritic branches compared to wild-type mice at P14, but not P7 [Citation149]. Also, mutant mice had approximately six times more neurons with dendritic arbors smaller than 600 um than their littermate wild-type mice [Citation149]. This change in dendritic length and branches was due to activity-dependent retraction of dendrites as seen in primary neurons transfected with Cav1.2-G406R channels and in TS patient iPSC-derived neurons [Citation149]. While the synaptic input frequency and strength to layer 2/3 excitatory neurons are unknown in mice with the G406R mutation, spontaneous calcium transients are increased in cortical neurons expressing Cav1.2-G406R channels during development [Citation131]. It is possible that the shorter dendritic length and branching observed at P14 [Citation149] is an accumulation of increased calcium transients that induced activity-dependent dendritic retraction. Interestingly, treating wild-type mice with LTCC blocker nimodipine at E12.5 or knocking out Cacna1c at E12.5 via in utero electroporation reduced neurite length [Citation131], suggesting that activity-dependent dendritic retraction seen as a result of TS-Cav1.2 may recruit alternate mechanisms.

Layer 5 cortical neurons in the adult prefrontal cortex of forebrain-specific Cacna1c conditional knockout mice (using CaMKIIa-Cre [Citation150]) show increased presynaptic excitatory and inhibitory inputs. Also, the relative protein level of VGLUT1 was higher than VGAT (more excitatory inputs than inhibitory inputs) [Citation150], which may suggest the net effect on pyramidal neurons is increased excitatory inputs. It is possible that increased excitatory inputs may lead to activity-dependent retraction of dendrites. Interestingly, treating with LTCC agonist FPL64176 marginally rescued the neurite reduction seen in Cacna1c KO condition, which is speculated to be via Cav1.3 stimulation [Citation131]. In fact, heterozygous loss of Cacna1d in new-born neurons of the hippocampus show shorter dendritic branch length [Citation130], suggesting that Cacna1d may be critical for dendritic branching during development in other brain regions. Together, these studies suggest that aberrant LTCC expression or channel function in developing cortical neurons impacts their dendritic morphology, which in turn will affect their communication with other brain regions.

Similar to dendritic morphology changes, axon projections are also altered with Cav1.2 LTCC mutations. In callosal projection neurons (neurons that make axonal projections from one side of the hemisphere to the other) with excessive WT Cav1.2 channels at E15.5 (when upper layer neurons are born), there is a reduction in axon arborization (i.e. spread of the axons) in layers 1–3 and layer 5 [Citation133]. Furthermore, Cav1.2 channels with the TS-like I1166T mutation reduced axon arborization in layers 1–3 and layer 5, but only layers 1–3 showed a stronger reduction compared to when overexpressing Cav1.2-WT channels [Citation133]. The reduced arborization was not due to a change in axon density in white matter, i.e., number of projections received from the contralateral hemisphere, suggesting that axon guidance during development is not perturbed in callosal projection neurons with excessive calcium influx. The aberrant axon arborization as a result of Cav1.2 WT and TS-like Cav1.2 I1166T mutation overexpression was partially rescued only in layers 1–3 when calcium permeability was reduced (L745P mutation) and Cav1.2-α1 and Cav1.2-β subunits (via W440A mutation) were blocked [Citation137]. However, it remains unknown whether the TS Cav1.2-G406R mutation or loss of Cacna1c or Cacna1d impacts axonogenesis.

Similar to neurons, in TS2-neo mice that express G406R mutant channels, oligodendrocyte progenitor cells (OPCs) show increased calcium influx at baseline and show higher extension of processes [Citation123]. Interestingly, unlike neurons that show decreased dendritic morphology, there were more OPCs that showed more complex morphology (more branching) in TS2-neo compared to control mice. This increased proportion of mature OPCs may underlie the increased myelination observed in TS2-neo mice in the corpus callosum [Citation123]. Additionally, the increase in myelination is not due to an increased number of OPCs or oligodendrocytes, as their numbers in TS2-neo mice were comparable to control mice. Whether the increase in myelination underlies social and cognitive behavioral deficits see in TS2 mice is unclear, increased myelination has been observed in ASD [Citation151–153] and seen in mice with ASD-like behavioral deficits [Citation154,Citation155]. In addition, the effect of loss of Cacna1c on myelination remains to be examined.

The role of LTCCs in synaptogenesis, axonogenesis, and myelination has only been studied in gain-of-function or loss-of-function in Cacna1c and not in Cacna1d. Three de novo mutations in CACNA1D have been associated with autism (p.A749G and p.G407R, see Pinggera et al., 2015 [Citation24]; S652L, see Hofer et al., 2020 [Citation156]), and these mutations in HEK293 cells (human embryonic kidney cells) show changes in their electrophysiological properties comparable to gain-of-function changes. Therefore, it is possible that introducing these Cacna1d mutations in vivo or in vitro in neurons may show similar changes in synaptogenesis and axonogenesis as Cacna1c gain-of-function studies have reported.

Cav1.2 and Cav1.3 outside of the cerebral cortex

Cacna1c and Cacna1d expression across development is not restricted to the cerebral cortex, but also the hippocampus and cerebellum, and all three brain regions show a similar developmental expression pattern [Citation128]. This suggests that mutations in Cacna1c or Cacna1d in the hippocampus or cerebellum may show similar deficits as the manipulations done in the cerebral cortex. In fact, loss of Cacna1c in cerebral cortical and hippocampal excitatory neurons (using Emx1IRES-Cre) leads to smaller cerebral cortex and hippocampal volume [Citation157], but it is unclear whether the change in volume is due to differences in cell number or dendritic arborization. As the thickness of the somatosensory cortex was not different [Citation157], it is likely that loss of Cacna1c affects dendritic arborization that may result in volumetric changes. Nevertheless, the exact role of Cacna1c and Cacna1d in the development of the hippocampus and cerebellum as well as other brain regions that may express these channels during development remains to be studied.

Timing of Cav1.2 or Cav1.3 mutations and behavioral consequences

Behavioral changes associated with Cav1.2 and Cav1.3 mutations may be dependent on the timing at which the mutation occurs during brain development and maturation. Embryonic deletion of Cacna1c in postmitotic forebrain glutamatergic neurons (Nex-Cre) promotes neuropsychiatric disorder-associated phenotypes, such as cognitive decline, impaired synaptic plasticity, reduced sociability, hyperactivity, and increased anxiety [Citation2]. Additionally, this embryonic deletion increases the susceptibility to chronic stress, suggesting that Cav1.2 interacts with the environment to shape later disease vulnerability [Citation2]. This is recapitulated in humans through the interaction of CACNA1C SNPs with adverse life events to later alter the risk of developing symptoms of a neuropsychiatric disorder, seen specifically in the interaction of CACNA1C and adult trauma to predict depression symptoms in humans [Citation2]. However, this phenotype is not seen when Cacna1c is deleted in glutamatergic neurons in adulthood (tamoxifen administration in postnatal weeks 11–13 in Camk2a-CreER line) [Citation2], suggesting that Cacna1c mutation-related behavioral changes are developmentally driven. Similarly, mice with heterozygous loss of Cacna1c show anxiety-like phenotype, impaired social behavior, learning, and memory [Citation150] but mice with Cacna1c deleted in adult forebrain glutamatergic neurons only exhibit anxiety-like phenotype and social deficits [Citation150,Citation158].

Conclusions

From the studies described in the genomic regulation section of this review, it is clear that neuropsychiatric disease-associated SNPs within non-coding regions of CACNA1C have the potential to impact gene expression by disrupting a variety of mechanisms that are normally recruited during transcription. These include (1) structural changes in chromatin (such as looping) and (2) epigenetic changes that include DNA methylation, histone modifications, regulation by microRNAs, and splicing. Thus, even though the significance of intronic SNPs to disease has been a question, the recent focus on understanding basic mechanisms of transcriptional regulation at the genomic level will allow us to probe how intronic SNPs (like those found in neuropsychiatric and neurodevelopmental disease-linked genes CACNA1C and CACNA1D) can impact gene expression. From the studies described in the brain development section, it is evident that alterations in either levels of Cav1.2 and Cav1.3 channels through altered gene expression or in function of these channels can impact certain aspects of neuronal development. Given the compelling evidence for a role for calcium signaling in brain development, the time is prime to further explore the role of Cav1.2 and Cav1.3 channels in brain development to connect the dots between genetic aberrations, impact on the brain, and disease-associated behaviors.

Acknowledgments

MRB is supported by the Graduate Research Fellowships Program (NSF213291) from the National Science Foundation. ASL is supported by Weill Cornell Medicine Clinical & Translational Science Center Predoctoral Training Award (TL1TR002386) from National Center for Advancing Translational Sciences. AMR is supported by NIH grants R01 MH118934 R01 DA050454, R01 MH125006, R01 DA029122 and the Weill Cornell Autism Research Program.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by the National Science Foundation [GRFP NSF213291]; National Institutes of Health [R01MH118934, R01DA050454, R01MH125006, R01DA053261, RO1DA054368]; National Center for Advancing Translation Sciences [TL1TR002386]; Weill Cornell Autism Research Program [0000].

References

  • Striessnig J, Pinggera A, Kaur G, et al. L-type Ca(2+) channels in heart and brain. Wiley Interdiscip Rev Membr Transp Signal. 2014 Mar;3(2):15–19.
  • Dedic N, Pohlmann ML, Richter JS, et al. Cross-disorder risk gene CACNA1C differentially modulates susceptibility to psychiatric disorders during development and adulthood. Mol Psychiatry. 2018 Mar;23(3):533–543.
  • Fleiss B, Rivkees SA, Gressens P. Early origins of neuropsychiatric disorders. Pediatr Res. 2019;85(2):113–114.
  • Taber KH, Jay Salpekar MD, Albert HC, et al. Developmental origins for neuropsychiatric illness. J Neuropsychiatry Clin Neurosci. 2011;23(1):1–5.
  • Faa G, Manchia M, Pintus R, et al. Fetal programming of neuropsychiatric disorders. Birth Defects Res C Embryo Today. 2016 Sep;108(3):207–223.
  • Splawski I, Timothy KW, Sharpe LM, et al. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell. 2004 Oct 1;119(1):19–31.
  • Splawski I, Timothy KW, Decher N, et al. Severe arrhythmia disorder caused by cardiac L-type calcium channel mutations. Proc Natl Acad Sci U S A. 2005 Jun 07;102(23):8089–8096. discussion 8086-8
  • Wemhöner K, Friedrich C, Stallmeyer B, et al. Gain-of-function mutations in the calcium channel CACNA1C (Cav1.2) cause non-syndromic long-QT but not Timothy syndrome. J Mol Cell Cardiol. 2015;80:186–195.
  • Marcantoni A, Calorio C, Hidisoglu E, et al. Cav1.2 channelopathies causing autism: new hallmarks on Timothy syndrome. Pflugers Arch. 2020 Jul;472(7):775–789.
  • Bamgboye MA, Herold KG, Vieira DCO, et al. CaV1.2 channelopathic mutations evoke diverse pathophysiological mechanisms. J Gen Physiol. 2022 Nov 7;154(11):e202213209.
  • Herold KG, Hussey JW, Dick IE. CACNA1C-Related Channelopathies. Handb Exp Pharmacol. 2023 Jan 5. doi:10.1007/164_2022_624. Online ahead of print.
  • Ortner NJ, Kaserer T, Copeland JN, et al. De novo CACNA1D Ca(2+) channelopathies: clinical phenotypes and molecular mechanism. Pflugers Arch. 2020 Jul;472(7):755–773.
  • Ortner NJ. CACNA1D-related channelopathies: from hypertension to autism. Handb Exp Pharmacol. 2023 Jan 3. doi:10.1007/164_2022_626. Online ahead of print.
  • Green EK, Grozeva D, Jones I, et al. The bipolar disorder risk allele at CACNA1C also confers risk of recurrent major depression and of schizophrenia. Mol Psychiatry. 2010 Oct;15(10):1016–1022.
  • Liu Y, Blackwood DH, Caesar S, et al. Meta-analysis of genome-wide association data of bipolar disorder and major depressive disorder. Mol Psychiatry. 2011;162011:2–4.
  • Group. PGCBDW. Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nat Genet. 2011 Sep 18;43(10):977–983.
  • Ament SA, Szelinger S, Glusman G, et al. Rare variants in neuronal excitability genes influence risk for bipolar disorder. Proc Natl Acad Sci U S A. 2015 Mar 17;112(11):3576–3581.
  • Nyegaard M, Demontis D, Foldager L, et al. CACNA1C (rs1006737) is associated with schizophrenia. Mol Psychiatry. 2010 Feb;15(2):119–121.
  • Rao S, Yao Y, Zheng C, et al. Common variants in CACNA1C and MDD susceptibility: a comprehensive meta-analysis. Am J Med Genet B Neuropsychiatr Genet. 2016 Sep;171(6):896–903.
  • Li J, Zhao L, You Y, et al. Schizophrenia related variants in CACNA1C also confer risk of autism. PLoS One. 2015;10(7):e0133247.
  • Consortium C-DGotPG. Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet. 2013 Apr 20;381(9875):1371–1379.
  • Kabir ZD, Martinez-Rivera A, Rajadhyaksha AM. From gene to behavior: l-type calcium channel mechanisms underlying neuropsychiatric symptoms. Neurotherapeutics. 2017 Jul;14(3):588–613.
  • Kabir ZD, Lee AS, Rajadhyaksha AM. L-type Ca(2+) channels in mood, cognition and addiction: integrating human and rodent studies with a focus on behavioural endophenotypes. J Physiol. 2016 Oct 15;594(20):5823–5837.
  • Pinggera A, Lieb A, Benedetti B, et al. CACNA1D de novo mutations in autism spectrum disorders activate Cav1.3 L-type calcium channels. Biol Psychiatry. 2015 May 1;77(9):816–822.
  • Zhang F, Lupski JR. Non-coding genetic variants in human disease. Hum Mol Genet. 2015 Oct 15;24(R1):R102–10.
  • Barešić A, Nash AJ, Dahoun T, et al. Understanding the genetics of neuropsychiatric disorders: the potential role of genomic regulatory blocks. Mol Psychiatry. 2020 Jan;25(1):6–18.
  • Schaub MA, Boyle AP, Kundaje A, et al. Linking disease associations with regulatory information in the human genome. Genome Res. 2012 Sep;22(9):1748–1759.
  • Maurano MT, Humbert R, Rynes E, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science. 2012 Sep 7;337(6099):1190–1195.
  • Pașca SP, Arlotta P, Bateup HS, et al. A nomenclature consensus for nervous system organoids and assembloids. Nature. 2022 Sep;609(7929):907–910.
  • Li M, Li T, Xiao X, et al. Phenotypes, mechanisms and therapeutics: insights from bipolar disorder GWAS findings. Mol Psychiatry. 2022 Jul;27(7):2927–2939.
  • Moon AL, Haan N, Wilkinson LS, et al. CACNA1C: association with psychiatric disorders, behavior, and neurogenesis. Schizophr Bull. 2018 Aug 20;44(5):958–965.
  • Shnayder NA, Novitsky MA, Neznanov NG, et al. Genetic predisposition to schizophrenia and depressive disorder comorbidity. Genes (Basel). 2022 Mar 2 ;13(3):457.
  • Heyes S, Pratt WS, Rees E, et al. Genetic disruption of voltage-gated calcium channels in psychiatric and neurological disorders. Prog Neurobiol. 2015;134:36–54.
  • Harrison PJ, Husain SM, Lee H, et al. CACNA1C (Ca(V)1.2) and other L-type calcium channels in the pathophysiology and treatment of psychiatric disorders: advances from functional genomics and pharmacoepidemiology. Neuropharmacology. 2022 Sep 22;220:109262.
  • Dekker J, Rippe K, Dekker M, et al. Capturing chromosome conformation. Science. 2002 Feb 15;295(5558):1306–1311.
  • Denker A, de Laat W. The second decade of 3C technologies: detailed insights into nuclear organization. Genes Dev. 2016 Jun 15;30(12):1357–1382.
  • Bickmore WA. The spatial organization of the human genome. Annu Rev Genomics Hum Genet. 2013;14:67–84.
  • Yu M, Ren B. The three-dimensional organization of mammalian genomes. Annu Rev Cell Dev Biol. 2017 Oct;6(33):265–289.
  • Kornberg RD. Chromatin structure: a repeating unit of histones and DNA. Science. 1974 May 24;184(4139):868–871.
  • MacAlpine DM, Almouzni G. Chromatin and DNA replication. Cold Spring Harb Perspect Biol. 2013 Aug 1;5(8):a010207.
  • Luger K, Mäder AW, Richmond RK, et al. Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature. 1997 Sep 18;389(6648):251–260.
  • Fujita Y, Pather SR, Ming GL, et al. 3D spatial genome organization in the nervous system: from development and plasticity to disease. Neuron. 2022;110(18): ;2902–2915.
  • Birney E, Stamatoyannopoulos JA, Dutta A, et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007 Jun 14;447(7146):799–816.
  • EP C. The ENCODE (ENCyclopedia Of DNA Elements) project. Science. 2004 Oct 22;306(5696):636–640.
  • Guo Y, Jamison DC. The distribution of SNPs in human gene regulatory regions. BMC Genomics. 2005 Oct;6(6):140.
  • Won H, de la Torre-Ubieta L, Stein JL, et al. Chromosome conformation elucidates regulatory relationships in developing human brain. Nature. 2016 Oct 27;538(7626):523–527.
  • Wray NR, Ripke S, Mattheisen M, et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat Genet. 2018 May;50(5):668–681.
  • Rajarajan P, Jiang Y, Kassim BS, et al. Chromosomal conformations and epigenomic regulation in schizophrenia. Prog Mol Biol Transl Sci. 2018;157:21–40.
  • Kadauke S, Blobel GA. Chromatin loops in gene regulation. Biochim Biophys Acta. 2009 Jan;1789(1):17–25.
  • Sadowski M, Kraft A, Szalaj P, et al. Spatial chromatin architecture alteration by structural variations in human genomes at the population scale. Genome Biol. 2019 Jul 30;20(1):148.
  • Behrends M, Engmann O. Loop interrupted: dysfunctional chromatin relations in neurological diseases. Front Genet. 2021;12:732033.
  • Grubert F, Srivas R, Spacek DV, et al. Landscape of cohesin-mediated chromatin loops in the human genome. Nature. 2020 Jul;583(7818):737–743.
  • Lee DP, Tan WLW, Anene-Nzelu CG, et al. Robust CTCF-based chromatin architecture underpins epigenetic changes in the heart failure stress–gene response. Circulation. 2019;139(16):1937–1956.
  • Roussos P, Mitchell AC, Voloudakis G, et al. A role for noncoding variation in schizophrenia. Cell Rep. 2014 Nov 20;9(4):1417–1429.
  • Mitchell AC, Bharadwaj R, Whittle C, et al. The genome in three dimensions: a new frontier in human brain research. Biol Psychiatry. 2014 Jun 15;75(12):961–969.
  • Lambert JT, Su-Feher L, Cichewicz K, et al. Parallel functional testing identifies enhancers active in early postnatal mouse brain. Elife. 2021;4;10:Oct.
  • Eckart N, Song Q, Yang R, et al. Functional characterization of schizophrenia-associated variation in CACNA1C. PLoS One. 2016;11(6):e0157086.
  • Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016 Aug;17(8):487–500.
  • Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell. 2007 Feb 23;128(4):635–638.
  • Kuehner JN, Bruggeman EC, Wen Z, et al. Epigenetic regulations in neuropsychiatric disorders. Front Genet. 2019;10:268.
  • Newell-Price J, Clark AJ, King P. DNA methylation and silencing of gene expression. Trends Endocrinol Metab. 2000 May-Jun;11(4):142–148.
  • Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity (Edinb). 2010 Jul;105(1):4–13.
  • Bird AP. DNA methylation versus gene expression. Development. 1984;83(Supplement):31–40.
  • Lev Maor G, Yearim A, Ast G. The alternative role of DNA methylation in splicing regulation. Trends Genet. 2015 May;31(5):274–280.
  • Maunakea AK, Chepelev I, Cui K, et al. Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res. 2013 Nov;23(11):1256–1269.
  • Shukla S, Kavak E, Gregory M, et al. CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing. Nature. 2011;479(7371):74–79.
  • Maunakea AK, Nagarajan RP, Bilenky M, et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 2010 Jul 8;466(7303):253–257.
  • Cheong J, Yamada Y, Yamashita R, et al. Diverse DNA methylation statuses at alternative promoters of human genes in various tissues. DNA Res. 2006;13(4):155–167.
  • Lyko F. The DNA methyltransferase family: a versatile toolkit for epigenetic regulation. Nat Rev Genet. 2018 Feb;19(2):81–92.
  • Weber M, Schübeler D. Genomic patterns of DNA methylation: targets and function of an epigenetic mark. Curr Opin Cell Biol. 2007 Jun;19(3):273–280.
  • Leonhardt H, Page AW, Weier HU, et al. A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell. 1992 Nov 27;71(5):865–873.
  • Okano M, Bell DW, Haber DA, et al. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999 Oct 29;99(3):247–257.
  • Saxena S, Maroju PA, Choudhury S, et al. Analysis of transcript levels of a few schizophrenia candidate genes in neurons from a transgenic mouse embryonic stem cell model overexpressing DNMT1. Gene. 2020 Oct 5;757:144934.
  • Vohra M, Sharma AR, Prabhu BN, et al. SNPs in sites for DNA methylation, transcription factor binding, and miRNA targets leading to allele-specific gene expression and contributing to complex disease risk: a systematic review. Public Health Genomics. 2020;23:155–170. Switzerland: © 2020S. Karger AG, Basel.
  • Starnawska A, Demontis D, Pen A, et al. CACNA1C hypermethylation is associated with bipolar disorder. Transl Psychiatry. 2016 Jun 7;6(6):e831.
  • Hannon E, Dempster EL, Mansell G, et al. DNA methylation meta-analysis reveals cellular alterations in psychosis and markers of treatment-resistant schizophrenia. Elife. 2021;26:Feb; 10.
  • Krzyzewska IM, Ensink JBM, Nawijn L, et al. Genetic variant in CACNA1C is associated with PTSD in traumatized police officers. Eur J Hum Genet. 2018 Feb;26(2):247–257.
  • Córdova-Palomera A, Fatjó-Vilas M, Gastó C, et al. Genome-wide methylation study on depression: differential methylation and variable methylation in monozygotic twins. Transl Psychiatry. 2015 Apr 28;5(4):e557.
  • Jaffe AE, Gao Y, Deep-Soboslay A, et al. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat Neurosci. 2016 Jan;19(1):40–47.
  • Zhu X, Li R, Kang G, et al. CACNA1C polymorphism (rs2283291) is associated with schizophrenia in Chinese males: a case-control study. Dis Markers. 2019;2019:8062397.
  • Zhang Y, Sun Z, Jia J, et al. Overview of histone modification. Adv Exp Med Biol. 2021;1283:1–16.
  • Huisinga KL, Brower-Toland B, Elgin SC. The contradictory definitions of heterochromatin: transcription and silencing. Chromosoma. 2006 Apr;115(2):110–122.
  • Babu A, Verma RS. Chromosome structure: euchromatin and heterochromatin. Int Rev Cytol. 1987;108:1–60.
  • Heintzman ND, Stuart RK, Hon G, et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet. 2007 Mar;39(3):311–318.
  • Creyghton MP, Cheng AW, Welstead GG, et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci U S A. 2010 Dec 14;107(50):21931–21936.
  • Fiorentino A, O’Brien NL, Locke DP, et al. Analysis of ANK3 and CACNA1C variants identified in bipolar disorder whole genome sequence data. Bipolar Disord. 2014;16(6):583–591.
  • Billingsley KJ, Manca M, Gianfrancesco O, et al. Regulatory characterisation of the schizophrenia-associated CACNA1C proximal promoter and the potential role for the transcription factor EZH2 in schizophrenia aetiology. Schizophr Res. 2018;199:168–175.
  • Zhao Z, Li Y, Chen H, et al. PD_NGSAtlas: a reference database combining next-generation sequencing epigenomic and transcriptomic data for psychiatric disorders. BMC Med Genomics. 2014 Dec;31(7):71.
  • Cannell IG, Kong YW, Bushell M. How do microRNAs regulate gene expression?. Biochem Soc Trans. 2008 Dec;36(Pt 6):1224–1231.
  • Selbach M, Schwanhäusser B, Thierfelder N, et al. Widespread changes in protein synthesis induced by microRNAs. Nature. 2008 Sep 4;455(7209):58–63.
  • Issler O, Chen A. Determining the role of microRNAs in psychiatric disorders. Nat Rev Neurosci. 2015 Apr;16(4):201–212.
  • Kyzar EJ, Bohnsack JP, Pandey SC. Current and future perspectives of noncoding RNAs in brain function and neuropsychiatric disease. Biol Psychiatry. 2022 Jan 15;91(2):183–193.
  • Guan F, Zhang B, Yan T, et al. MIR137 gene and target gene CACNA1C of miR-137 contribute to schizophrenia susceptibility in Han Chinese. Schizophr Res. 2014 Jan;152(1):97–104.
  • Kwon E, Wang W, Tsai LH. Validation of schizophrenia-associated genes CSMD1, C10orf26, CACNA1C and TCF4 as miR-137 targets. Mol Psychiatry. 2013 Jan ;18(1):11–12.
  • Consortium SPG-WASG. Genome-wide association study identifies five new schizophrenia loci. Nat Genet. 2011 Sep 18;43(10):969–976.
  • Collins AL, Kim Y, Bloom RJ, et al. Transcriptional targets of the schizophrenia risk gene MIR137. Transl Psychiatry. 2014 Jul 1;4(7):e404.
  • Gu X, Xu Y, Xue WZ, et al. Interplay of miR-137 and EZH2 contributes to the genome-wide redistribution of H3K27me3 underlying the Pb-induced memory impairment. Cell Death Dis. 2019 Sep 11;10(9):671.
  • Wright C, Turner JA, Calhoun VD, et al. Potential Impact of miR-137 and Its Targets in Schizophrenia. Front Genet. 2013;4:58.
  • Szulwach KE, Li X, Smrt RD, et al. Cross talk between microRNA and epigenetic regulation in adult neurogenesis. J Cell Biol. 2010 Apr 5;189(1):127–141.
  • Lee AS, De Jesus-Cortes H, Kabir ZD, et al. The neuropsychiatric disease-associated gene cacna1c mediates survival of young hippocampal neurons. eNeuro. 2016 Mar 31;3(2):ENEURO.0006–16.2016.
  • Tartt AN, Mariani MB, Hen R, et al. Dysregulation of adult hippocampal neuroplasticity in major depression: pathogenesis and therapeutic implications. Mol Psychiatry. 2022 Jun;27(6):2689–2699.
  • Anacker C, Hen R. Adult hippocampal neurogenesis and cognitive flexibility - linking memory and mood. Nat Rev Neurosci. 2017 Jun;18(6):335–346.
  • Kang E, Wen Z, Song H, et al. Adult neurogenesis and psychiatric disorders. Cold Spring Harb Perspect Biol. 2016 1;8(9).
  • He E, Lozano MAG, Stringer S, et al. MIR137 schizophrenia-associated locus controls synaptic function by regulating synaptogenesis, synapse maturation and synaptic transmission. Hum Mol Genet. 2018 Jun 1;27(11):1879–1891.
  • Yoshikawa A, Kushima I, Miyashita M, et al. Dysregulation of post-transcriptional modification by copy number variable microRNAs in schizophrenia with enhanced glycation stress. Transl Psychiatry. 2021 May 28;11(1):331.
  • Nilsen TW, Graveley BR. Expansion of the eukaryotic proteome by alternative splicing. Nature. 2010 Jan 28;463(7280):457–463.
  • Glatt SJ, Cohen OS, Faraone SV, et al. Dysfunctional gene splicing as a potential contributor to neuropsychiatric disorders. Am J Med Genet B Neuropsychiatr Genet. 2011 Jun;156b(4):382–392.
  • Flaherty E, Zhu S, Barretto N, et al. Neuronal impact of patient-specific aberrant NRXN1α splicing. Nat Genet. 2019 Dec;51(12):1679–1690.
  • Latorre E, Mesonero JE, Harries LW. Alternative splicing in serotonergic system: implications in neuropsychiatric disorders. J Psychopharmacol. 2019 Nov;33(11):1352–1363.
  • Carvill GL, Mefford HC. Poison exons in neurodevelopment and disease. Curr Opin Genet Dev. 2020 Dec;65:98–102.
  • Porter RS, Jaamour F, Iwase S. Neuron-specific alternative splicing of transcriptional machineries: implications for neurodevelopmental disorders. Mol Cell Neurosci. 2018 Mar;87:35–45.
  • Zhou H-L, Luo G, Wise JA, et al. Regulation of alternative splicing by local histone modifications: potential roles for RNA-guided mechanisms. Nucleic Acids Res. 2013;42(2):701–713.
  • Mucaki EJ, Shirley BC, Rogan PK. Expression changes confirm genomic variants predicted to result in allele-specific, alternative mRNA splicing. Front Genet. 2020;11:109.
  • Monlong J, Calvo M, Ferreira PG, et al. Identification of genetic variants associated with alternative splicing using sQTLseekeR. Nat Commun. 2014 Aug;20(5):4698.
  • Clark MB, Wrzesinski T, Garcia AB, et al. Long-read sequencing reveals the complex splicing profile of the psychiatric risk gene CACNA1C in human brain. Mol Psychiatry. 2020 Jan;25(1):37–47.
  • Panagiotakos G, Haveles C, Arjun A, et al. Aberrant calcium channel splicing drives defects in cortical differentiation in Timothy syndrome. Elife. 2019;23:Dec; 8.
  • Banono NS, Gawel K, De Witte L, et al. Zebrafish larvae carrying a splice variant mutation in cacna1d: a new model for schizophrenia-like behaviours?. Mol Neurobiol. 2021 Feb;58(2):877–894.
  • Kandel ER, Schwartz JH, Jessell TM. Principles of Neural Science. 4th ed. McGraw-Hill: Health Professions Division; 2000.
  • Valiente M, Marín O. Neuronal migration mechanisms in development and disease. Curr Opin Neurobiol. 2010 Feb;20(1):68–78.
  • Manzini MC, Walsh CA. What disorders of cortical development tell us about the cortex: one plus one does not always make two. Curr Opin Genet Dev. 2011 Jun;21(3):333–339.
  • Rubenstein JL. Annual research review: development of the cerebral cortex: implications for neurodevelopmental disorders. J Child Psychol Psychiatry. 2011 Apr;52(4):339–355.
  • Pasca SP, Portmann T, Voineagu I, et al. Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat Med. 2011 Dec;17(12):1657–1662.
  • Cheli VT, Santiago González DA, Zamora NN, et al. Enhanced oligodendrocyte maturation and myelination in a mouse model of Timothy syndrome. Glia. 2018 Nov;66(11):2324–2339.
  • Horigane SI, Ozawa Y, Zhang J, et al. A mouse model of Timothy syndrome exhibits altered social competitive dominance and inhibitory neuron development. FEBS Open Bio. 2020 Aug;10(8):1436–1446.
  • Temme SJ, Bell RZ, Fisher GL, et al. Deletion of the mouse homolog of CACNA1C disrupts discrete forms of hippocampal-dependent memory and neurogenesis within the dentate gyrus. eNeuro. 2016 Nov 28;3(6): ENEURO.0118–16.2016.
  • Völkening B, Schönig K, Kronenberg G, et al. Deletion of psychiatric risk gene Cacna1c impairs hippocampal neurogenesis in cell-autonomous fashion. Glia. 2017 May;65(5):817–827.
  • Redecker TM, Kisko TM, Wöhr M, et al. Cacna1c haploinsufficiency lacks effects on adult hippocampal neurogenesis and volumetric properties of prefrontal cortex and hippocampus in female rats. Physiol Behav. 2020 Sep 1;223:112974.
  • Schlick B, Flucher BE, Obermair GJ. Voltage-activated calcium channel expression profiles in mouse brain and cultured hippocampal neurons. Neuroscience. 2010 May 19;167(3):786–798.
  • Marschallinger J, Sah A, Schmuckermair C, et al. The L-type calcium channel Cav1.3 is required for proper hippocampal neurogenesis and cognitive functions. Cell Calcium. 2015 Dec;58(6):606–616.
  • Kim SH, Park YR, Lee B, et al. Reduction of Cav1.3 channels in dorsal hippocampus impairs the development of dentate gyrus newborn neurons and hippocampal-dependent memory tasks. PLoS One. 2017;12(7):e0181138.
  • Kamijo S, Ishii Y, Horigane SI, et al. A critical neurodevelopmental role for L-type voltage-gated calcium channels in neurite extension and radial migration. J Neurosci. 2018 Jun 13;38(24):5551–5566.
  • Horigane SI, Hamada S, Kamijo S, et al. Development of an L-type Ca(2+) channel-dependent Ca(2+) transient during the radial migration of cortical excitatory neurons. Neurosci Res. 2021;169:17–26.
  • Nakagawa-Tamagawa N, Kirino E, Sugao K, et al. Involvement of calcium-dependent pathway and β subunit-interaction in neuronal migration and callosal projection deficits caused by the Cav1.2 I1166T mutation in developing mouse neocortex. Front Neurosci. 2021;15:747951.
  • Birey F, Andersen J, Makinson CD, et al. Assembly of functionally integrated human forebrain spheroids. Nature. 2017 May 4;545(7652):54–59.
  • Birey F, Pașca SP. Imaging neuronal migration and network activity in human forebrain assembloids. STAR Protoc. 2022 Sep 16;3(3):101478.
  • Bader PL, Faizi M, Kim LH, et al. Mouse model of Timothy syndrome recapitulates triad of autistic traits. Proc Natl Acad Sci U S A. 2011 Sep 13;108(37):15432–15437.
  • Lee SM, Yeh PWL. Yeh HH. L-type calcium channels contribute to ethanol-induced aberrant tangential migration of primordial cortical GABAergic interneurons in the embryonic medial prefrontal cortex. eNeuro. 2022 Jan 28;9(1): ENEURO.0359–21.2021.
  • Molyneaux BJ, Arlotta P, Menezes JR, et al. Neuronal subtype specification in the cerebral cortex. Nat Rev Neurosci. 2007 Jun;8(6):427–437.
  • Leone DP, Srinivasan K, Chen B, et al. The determination of projection neuron identity in the developing cerebral cortex. Curr Opin Neurobiol. 2008 Feb;18(1):28–35.
  • Fame RM, MacDonald JL, Macklis JD. Development, specification, and diversity of callosal projection neurons. Trends Neurosci. 2011 Jan;34(1):41–50.
  • Srinivasan K, Leone DP, Bateson RK, et al. A network of genetic repression and derepression specifies projection fates in the developing neocortex. Proc Natl Acad Sci U S A. 2012 Nov 20;109(47):19071–19078.
  • Arlotta P, Molyneaux BJ, Chen J, et al. Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo. Neuron. 2005 Jan 20;45(2):207–221.
  • Chen B, Schaevitz LR, McConnell SK. Fezl regulates the differentiation and axon targeting of layer 5 subcortical projection neurons in cerebral cortex. Proc Natl Acad Sci U S A. 2005 Nov 22;102(47):17184–17189.
  • Molyneaux BJ, Arlotta P, Hirata T, et al. Fezl is required for the birth and specification of corticospinal motor neurons. Neuron. 2005 Sep 15;47(6):817–831.
  • Alcamo EA, Chirivella L, Dautzenberg M, et al. Satb2 regulates callosal projection neuron identity in the developing cerebral cortex. Neuron. 2008 Feb 7;57(3):364–377.
  • Arlotta P, Molyneaux BJ, Jabaudon D, et al. Ctip2 controls the differentiation of medium spiny neurons and the establishment of the cellular architecture of the striatum. J Neurosci. 2008 Jan 16;28(3):622–632.
  • Britanova O, de Juan Romero C, Cheung A, et al. Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex. Neuron. 2008 Feb 7;57(3):378–392.
  • Chen B, Wang SS, Hattox AM, et al. The Fezf2-Ctip2 genetic pathway regulates the fate choice of subcortical projection neurons in the developing cerebral cortex. Proc Natl Acad Sci U S A. 2008 Aug 12;105(32):11382–11387.
  • Krey JF, Paşca SP, Shcheglovitov A, et al. Timothy syndrome is associated with activity-dependent dendritic retraction in rodent and human neurons. Nat Neurosci. 2013 Feb;16(2):201–209.
  • Kabir ZD, Che A, Fischer DK, et al. Rescue of impaired sociability and anxiety-like behavior in adult cacna1c-deficient mice by pharmacologically targeting eIF2α. Mol Psychiatry. 2017 Aug;22(8):1096–1109.
  • Carmody DP, Lewis M. Regional white matter development in children with autism spectrum disorders. Dev Psychobiol. 2010 Dec;52(8):755–763.
  • Gozzi M, Nielson DM, Lenroot RK, et al. A magnetization transfer imaging study of corpus callosum myelination in young children with autism. Biol Psychiatry. 2012 Aug 1;72(3):215–220.
  • Broek JA, Guest PC, Rahmoune H, et al. Proteomic analysis of post mortem brain tissue from autism patients: evidence for opposite changes in prefrontal cortex and cerebellum in synaptic connectivity-related proteins. Mol Autism. 2014;5:41.
  • Khanbabaei M, Hughes E, Ellegood J, et al. Precocious myelination in a mouse model of autism. Transl Psychiatry. 2019 Oct 7;9(1):251.
  • Vacher CM, Lacaille H, O’Reilly JJ, et al. Placental endocrine function shapes cerebellar development and social behavior. Nat Neurosci. 2021 Oct;24(10):1392–1401.
  • Hofer NT, Tuluc P, Ortner NJ, et al. Biophysical classification of a CACNA1D de novo mutation as a high-risk mutation for a severe neurodevelopmental disorder. Mol Autism. 2020;11(1):4.
  • Smedler E, Louhivuori L, Romanov RA, et al. Disrupted Cacna1c gene expression perturbs spontaneous Ca(2+) activity causing abnormal brain development and increased anxiety. Proc Natl Acad Sci U S A. 2022 Feb 15;119:7.
  • Lee AS, Ra S, Rajadhyaksha AM, et al. Forebrain elimination of cacna1c mediates anxiety-like behavior in mice. Mol Psychiatry. 2012;172012:1054–1055.