Calcium influx can be stimulated by various intra- and extracellular signals to set coordinated gene expression programs into motion. As such, the precise regulation of intracellular calcium represents a nexus between environmental cues and intrinsic genetic programs. Mounting genetic evidence points to a role for the deregulation of intracellular calcium signaling in neuropsychiatric disorders of developmental origin. These findings have prompted renewed enthusiasm for understanding the roles of calcium during normal and dysfunctional prenatal development. In this Review, we describe the fundamental mechanisms through which calcium is spatiotemporally regulated and directs early neurodevelopmental events. We also discuss unanswered questions about intracellular calcium regulation during the emergence of neurodevelopmental disease, and provide evidence that disruption of cell-specific calcium homeostasis and/or redeployment of developmental calcium signaling mechanisms may contribute to adult neurological disorders. We propose that understanding the normal developmental events that build the nervous system will rely on gaining insights into cell type-specific calcium signaling mechanisms. Such an understanding will enable therapeutic strategies targeting calcium-dependent mechanisms to mitigate disease.

Calcium, which lies at the hub of multiple signal transduction pathways, is uniquely situated to transduce dynamic biological inputs into distinct cell behaviors (reviewed by Berridge et al., 2003). Calcium influx in the embryonic brain occurs in response to multiple developmental signals, including electrical activity, and dynamic elevations in cytoplasmic calcium are linked to transcriptional programs that are crucial for development, homeostasis and plasticity (reviewed by Greer and Greenberg, 2008; Lyons and West, 2011). In this Review, we present evidence supporting the idea that spatiotemporally regulated and cell-specific functions of calcium signaling transducers underlie the earliest cellular behaviors that build the cerebral cortex. We focus on calcium entry in developing cortical cells, incorporating lessons gleaned from other developing neural populations. We also detail cell biological outputs influenced by the precise control of cytoplasmic calcium, placing special emphasis on stem/progenitor populations and immature neuroblasts. Moreover, we discuss genetic evidence suggesting that deregulation of intracellular calcium represents a potential node of convergence for neurodevelopmental disorders.

Calcium signaling involves both the sensing and transduction of extracellular calcium (Brown et al., 1993), as well as the transport of calcium across the plasma membrane (reviewed by Berridge et al., 2003). Homeostatic tuning of intracellular calcium employs an extensive repertoire of cell surface and organellar ion channels, transporters, pumps and buffers (Fig. 1, Box 1), each possessing distinctive properties (e.g. selectivity, conductance, calcium affinity, kinetics). These effectively stage a dynamic process of intracellular compartmentalization, wherein calcium abundance in different subcellular compartments (e.g. cytoplasm, intraorganellar calcium stores) changes across space and time. Movement of calcium into and out of these compartments enables activation of distinct sets of calcium-sensitive proteins, based on their levels and localization (reviewed by Berridge et al., 2003; Clapham, 2007). How distributed networks of calcium signaling proteins confer signaling specificity in various cellular contexts remains a fundamental question. Below, we briefly describe mechanisms of calcium regulation in developing neural cells. We emphasize spatiotemporal properties of calcium signals, alternative splicing of genes regulating calcium signaling, and cell type-specific expression of calcium-dependent effectors. Coupled with the cellular niche and environmental signals to which a cell is exposed, the intersection of these factors influences calcium dynamics, ultimately dictating the selective activation of gene expression programs (reviewed by Clapham, 2007; Rosenberg and Spitzer, 2011).

Box 1. Key mechanisms of calcium entry in developing neural cells

An overview of intracellular calcium signaling is depicted in Fig. 1. Here, we focus on channels mediating two types of calcium entry in immature neural cells that are dependent on environmental signals and result in downstream calcium-dependent signaling and transcription.

Voltage-gated calcium channels (VGCCs) are a primary route of calcium influx in developing neural cells (Fig. 1) in response to neural activity. By converting changes in membrane potential at the cell surface into intracellular calcium elevations, VGCCs couple electrical activity to cell biological processes, including phosphorylation and transcription (reviewed by Catterall, 2011; Zamponi et al., 2015). VGCCs consist of a pore-forming subunit, which allows calcium entry in response to depolarizing stimuli, and auxiliary subunits regulating channel localization and function (reviewed by Catterall, 2011; Zamponi et al., 2015). VGCCs are divided into families based on their physiological properties, and different channels display unique properties and localization. This enables distinct roles for VGCCs in different tissues and particular combinations of deficits associated with specific VGCC mutations (reviewed by Catterall, 2011; Zamponi et al., 2015).

Another major source of calcium influx is store operated calcium entry (SOCE), which is activated upon endoplasmic reticulum (ER) calcium store depletion in response to extracellular signals (Fig. 1) (reviewed by Prakriya and Lewis, 2015). SOCE is mediated by ORAI plasma membrane channels (ORAI1-3, also referred to as calcium release-activated calcium, or CRAC, channels) and the stromal interaction molecule (STIM) family of ER calcium sensors (reviewed by Putney et al., 2017). Activation of plasma membrane receptor tyrosine kinases or G-protein coupled receptors by extracellular ligands promotes inositol trisphosphate (IP3)- or ryanodine (Ry)-mediated release of calcium into the cytosol from the ER lumen (reviewed by Prakriya and Lewis, 2015). When intraluminal calcium is depleted, STIM proteins on the ER membrane oligomerize and translocate to ER-plasma membrane junctions, trapping and interacting with ORAI channels to modulate calcium influx (reviewed by Prakriya and Lewis, 2015). SOCE through ORAI channels can generate distinct patterns of intracellular calcium fluctuations to regulate various molecular events, including transcriptional activation (reviewed by Lewis, 2011).

Fig. 1.

Overview of intracellular calcium signaling. Schematic highlighting ion channels and pumps mediating cytoplasmic calcium homeostasis and select calcium-dependent pathways that transduce calcium signals to the nucleus. Low resting cytosolic calcium levels are maintained by plasma membrane calcium ATPases (PMCA1-4, encoded by ATP2B1-4), which extrude calcium out of the cell and display low calcium efflux capacity but high calcium affinity, and pumps on the surface of the sarco/endoplasmic reticulum (SR/ER) and mitochondria (e.g., the SR/ER calcium ATPases SERCA1-3, encoded by ATP2A1-3), which transport calcium into intraorganellar stores. Large calcium elevations are countered by sodium calcium exchangers (NCX1-4, encoded by SLC8A1-4) on the plasma membrane and mitochondrial and ER membranes. Calcium-permeable channels mediating calcium influx from the extracellular space include voltage-gated calcium channels (VGCCs), N-methyl-D-aspartate receptors (NMDARs), α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors (AMPARs), transient receptor potential (TRP) channels and ORAI channels. VGCCs are activated by membrane depolarization, whereas ORAI channels allow calcium influx upon ER calcium store depletion. Successive release of calcium from the ER, mediated by IP3R or RyR, depletes ER calcium, which in turn activates STIM calcium sensors (STIM1 and STIM2) to promote their interaction with ORAI channels (ORAI 1-3) and subsequent calcium influx. Gap junctions in the plasma membrane allow for intercellular propagation of calcium signals, through direct transfer of ions or small molecules that are crucial for intracellular signaling (e.g. IP3). Calcium-sensitive proteins in the cytoplasm (e.g. calmodulin; CaM) undergo a conformational change upon calcium binding, initiating distinct signaling cascades to the nucleus that culminate in calcium-dependent transcription. Note that only select calcium-dependent transcription factors with reported roles in the developing nervous system are highlighted; other factors and organellar calcium stores (e.g. mitochondria, lysosomes, Golgi apparatus) are not depicted.

Fig. 1.

Overview of intracellular calcium signaling. Schematic highlighting ion channels and pumps mediating cytoplasmic calcium homeostasis and select calcium-dependent pathways that transduce calcium signals to the nucleus. Low resting cytosolic calcium levels are maintained by plasma membrane calcium ATPases (PMCA1-4, encoded by ATP2B1-4), which extrude calcium out of the cell and display low calcium efflux capacity but high calcium affinity, and pumps on the surface of the sarco/endoplasmic reticulum (SR/ER) and mitochondria (e.g., the SR/ER calcium ATPases SERCA1-3, encoded by ATP2A1-3), which transport calcium into intraorganellar stores. Large calcium elevations are countered by sodium calcium exchangers (NCX1-4, encoded by SLC8A1-4) on the plasma membrane and mitochondrial and ER membranes. Calcium-permeable channels mediating calcium influx from the extracellular space include voltage-gated calcium channels (VGCCs), N-methyl-D-aspartate receptors (NMDARs), α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors (AMPARs), transient receptor potential (TRP) channels and ORAI channels. VGCCs are activated by membrane depolarization, whereas ORAI channels allow calcium influx upon ER calcium store depletion. Successive release of calcium from the ER, mediated by IP3R or RyR, depletes ER calcium, which in turn activates STIM calcium sensors (STIM1 and STIM2) to promote their interaction with ORAI channels (ORAI 1-3) and subsequent calcium influx. Gap junctions in the plasma membrane allow for intercellular propagation of calcium signals, through direct transfer of ions or small molecules that are crucial for intracellular signaling (e.g. IP3). Calcium-sensitive proteins in the cytoplasm (e.g. calmodulin; CaM) undergo a conformational change upon calcium binding, initiating distinct signaling cascades to the nucleus that culminate in calcium-dependent transcription. Note that only select calcium-dependent transcription factors with reported roles in the developing nervous system are highlighted; other factors and organellar calcium stores (e.g. mitochondria, lysosomes, Golgi apparatus) are not depicted.

Spatiotemporal regulation of calcium signals

Calcium influx through different calcium-permeable channels engages distinct signaling cascades (Fig. 1), in part based on the localization of calcium-sensitive signaling proteins (Bading et al., 1993; Graef et al., 1999; reviewed by Parekh, 2008; West et al., 2001). Cytoplasmic calcium chelators, as well as ion pumps and exchangers that bind or extrude calcium (Fig. 1), create transient spatially-restricted high-calcium microenvironments within cells (reviewed by Parekh, 2008). This enables distinct contributions of local and global calcium elevations to intracellular signaling. Functional knock-in experiments and the use of calcium chelators with different affinities have revealed, for example, that local calcium elevations through L-type voltage-gated calcium channels (VGCCs) are transduced to the nucleus via a shuttle protein to promote activation of the transcription factor CREB (Deisseroth et al., 1996; Dolmetsch et al., 2001; Ma et al., 2014). In hippocampal neurons, it has been suggested that the calcium/calmodulin-activated phosphatase, calcineurin (CaN), is tethered to L-type VGCCs via an anchoring protein to contribute to the regulation of calcium influx through these channels (Oliveria et al., 2007, 2012).

Local translation of calcium signaling components can also influence calcium dynamics in specific cellular compartments, such as neuronal dendrites (Sun et al., 2021; reviewed by Holt et al., 2019). Moreover, emerging data reveal that calcium elevations near different intracellular calcium stores (e.g. lysosomes) regulate processes such as autophagy (Medina et al., 2015). In dendrites, endoplasmic reticulum (ER) stores participate in localized calcium signaling that is crucial for synaptic plasticity (Hirabayashi et al., 2017; O'Hare et al., 2022; Takechi et al., 1998). Mitochondrial calcium uptake, regulated by distinct mitochondrial morphologies in different neuronal compartments, can also modulate cytosolic calcium levels and processes such as neurotransmission (Lewis et al., 2018). Notably, spatial regulation of calcium is not restricted to individual cells, as propagation of calcium signals can occur across cohorts of coupled cells in the developing brain (Weissman et al., 2004).

Dynamic patterns of calcium fluctuations also enable signaling specificity, encoding information that is translated into long-lasting biochemical changes by cytosolic proteins with different calcium sensitivities (reviewed by Rosenberg and Spitzer, 2011). In T cells, for example, different patterns of calcium oscillations control transcriptional specificity (reviewed by Dolmetsch et al., 1997, 1998). Seminal studies of the developing Xenopus nervous system identified two types of spontaneous calcium transients contributing to distinct aspects of spinal neuron differentiation (Gomez and Spitzer, 1999; Gu and Spitzer, 1995; Gu et al., 1994). Recently, temporal waves of gene expression have been identified in cortical neurons, resulting from activity patterns of different durations (Tyssowski et al., 2018). The initial wave of early-response transcription factors induced by prolonged depolarization, including NPAS4 (Lin et al., 2008), corresponds to activity-dependent programs activated by brief stimulation. Other studies implicate Nuclear Factor of Activated T-cells (NFAT) transcription factors, previously linked to calcium influx through L-type VGCCs (Graef et al., 1999), as somatic calcium spike counters transducing dendritic VGCC activation to the nucleus (Wild et al., 2019). Dissecting how cell type-specific calcium transients encode information in the embryonic cortex will inform how these dynamics drive cell biological changes to impact developmental programs.

Coordination of routes of calcium entry

VGCCs and store-operated calcium entry (SOCE) are two major sources of calcium influx in developing neural cells (Box 1) (reviewed by Toth et al., 2016). Growing evidence indicates that these parallel entry mechanisms are reciprocally regulated (Park et al., 2010; Wang et al., 2010). In cortical neurons and vascular smooth muscle cells, the ER membrane protein STIM1 (Fig. 1), which activates ORAI channels to promote SOCE, attenuates calcium entry through the L-type VGCC Cav1.2 (Park et al., 2010; Wang et al., 2010). In the reverse direction, changes in membrane potential can alter calcium conductance through ORAI channels (Bakowski and Parekh, 2000). This bidirectional regulation may enable preferential use of store-operated or activity-dependent calcium entry in different cell types. In collaboration with calcium buffers, pumps and transporters, such crosstalk likely tunes calcium dynamics to control intracellular signaling. Much remains to be understood, however, about mechanisms coordinating calcium dynamics in the developing cortex to preferentially activate specific downstream transcriptional pathways.

Alternative splicing regulates calcium entry and homeostasis

Differentiation in the embryonic cortex is accompanied by global splicing changes (Zhang et al., 2016). Moreover, in different cell and tissue types, alternative splicing of calcium channels and calcium-dependent effectors regulates calcium signaling by generating functionally diverse isoforms (reviewed by Lipscombe et al., 2013). Precise isoform utilization in the embryonic cortex may, therefore, be an important contributor to cell- and region-specific calcium responses to developmental stimuli.

Alternative splicing of VGCC transcripts yields channel variants with specialized roles in different tissues, cells and cellular compartments (reviewed by Abernethy and Soldatov, 2002; Lipscombe et al., 2013). CACNA1C (encoding the pore-forming subunit of the VGCC Cav1.2), for example, is extensively spliced to generate channels with distinct properties (Soldatov, 1994; Soldatov et al., 1997; Tang et al., 2004), and long-read sequencing confirms region-specific CACNA1C splicing in the adult human brain (Clark et al., 2020). Stereotyped use of specific exons also yields dominant tissue-specific Cacna1c isoforms, suggesting the presence of coordinated splicing events (Tang et al., 2007; Welling et al., 1997). Disease-causing mutations restricted to specific isoforms could thus give rise to channelopathies reflecting their expression pattern (reviewed by Abernethy and Soldatov, 2002; Lipscombe et al., 2013).

Calcium channel splicing is also temporally regulated. Our studies and others point to developmental switches in CACNA1C/Cacna1c exon use in the brain, implying roles for different channel isoforms over time (Panagiotakos et al., 2019; Tang et al., 2009, 2011). For example, use of two mutually exclusive CACNA1C exons (8 and 8a), which are mutated in the syndromic autism spectrum disorder (ASD) Timothy Syndrome (TS), is developmentally controlled (Panagiotakos et al., 2019; Tang et al., 2011). In addition to impeding channel inactivation (Splawski et al., 2004), the TS mutation in exon 8a prevents a normal developmental splicing switch in patient cells (Table S1). This results in continued mutant exon inclusion in developing neurons and ensuing cellular phenotypes contributing to TS (Birey et al., 2017, 2022; Panagiotakos et al., 2019; Paşca et al., 2011; Table S1, discussed in more detail later). How channel subunit isoforms associate with one another, and how their expression is regulated across time, is unclear but likely influences cell type-specific calcium responses.

Tissue- and maturation state-specific PMCA (Fig. 1) isoform expression has also been reported (Brandt and Neve, 1992; Kip et al., 2006). Both neural activity and calcium regulate PMCA splicing (Carafoli et al., 1999; Zacharias and Strehler, 1996), supporting the idea that activity-dependent feedback influences calcium signaling via splicing regulation. Splice variants of many regulators of calcium homeostasis have been identified, including N-methyl-D-aspartate receptors (NMDARs) (An and Grabowski, 2007; Vallano et al., 1999), ORAI1 (Fukushima et al., 2012), STIM1 (Darbellay et al., 2011; Ramesh et al., 2021) and calcium-dependent transcription factors like NFAT (Vihma et al., 2008) and CREB (Walker et al., 1996). Mapping the cell type-specific expression of calcium channel and signaling protein isoforms across development is essential to understanding how calcium elicits specific responses in the embryonic cortex.

Mutations in genes impinging on calcium signaling have been implicated in neuropsychiatric disorders of developmental origin. In this section, we discuss genetic evidence highlighting that disrupted calcium-dependent molecular networks may contribute to misregulation of cellular behaviors in the developing brain. To aid our discussion, we surveyed the Simons Foundation Autism Research Initiative (SFARI) Gene database for studies implicating regulators of calcium signaling in the etiology of neurodevelopmental disorders. We restrict our analysis (summarized in Table S1 and discussed below) to genes encoding proteins that either: (1) directly regulate calcium entry, signaling or homeostasis, or (2) indirectly modulate calcium signaling by altering membrane potential or excitability. Due to space constraints, we have not included synaptic structural proteins that regulate ion/calcium channel localization and function at synapses [e.g. ANK2 (Kline et al., 2014), NRXN1-3 (Luo et al., 2020; Missler et al., 2003)], which have been reproducibly implicated in ASD.

Genetic studies have associated mutations in VGCC subunits with increased risk for neuropsychiatric disorders (Table S1). For example, CACNA1C variants are associated with bipolar disorder, schizophrenia and ASD (Cross-Disorder Group of the Psychiatric Genomics Consortium, 2013; reviewed by Bhat et al., 2012). Classical TS, a syndromic ASD, is caused by a point mutation in CACNA1C (Table S1). This mutation impairs both voltage-dependent Cav1.2 channel inactivation, resulting in elevated depolarization-induced calcium, and channel splicing, leading to persistent mutant channel expression in immature neurons (Panagiotakos et al., 2019; Paşca et al., 2011; Splawski et al., 2004). The ensuing abnormalities in channel signaling yield a constellation of cellular phenotypes, including differentiation deficits, activity-dependent dendritic retraction and impaired interneuron migration (Birey et al., 2017; Krey et al., 2013; Panagiotakos et al., 2019; Paşca et al., 2011). Mutations in genes encoding other calcium-permeable channels, such as GluN2 NMDAR subunits, have also been linked to neurodevelopmental conditions (Endele et al., 2010) (Table S1). In addition, variants of ATP2B2 (Iossifov et al., 2014), which encodes the PMCA2 calcium pump, and missense mutations in ITPR1, which encodes the IP3 receptor type 1 (IP3R1), are implicated in ASD (De Rubeis et al., 2014; Iossifov et al., 2014; Wang et al., 2016). In line with this, fibroblasts from individuals with syndromic and sporadic ASD display attenuated IP3-dependent calcium signaling (Schmunk et al., 2015, 2017).

Mutations in other ion channels and neurotransmitter receptors, which can indirectly influence voltage-dependent calcium influx, are also associated with neurodevelopmental disorders. Genetic variants in SCN2A, encoding the Nav1.2 sodium channel, for example, are strongly linked to infantile epilepsy, ASD and intellectual disability (ID) (reviewed by Sanders et al., 2018) (Table S1). One such mutation (K1422E) in Nav1.2 channels renders them calcium-permeable. Scn2aK1442E/+ cortical neurons display larger action potential-evoked calcium transients compared with wild-type neurons, suggesting that this mutation impacts calcium signaling (Echevarria-Cooper et al., 2022). Similarly, potassium channel mutations that alter excitability (e.g. KCNQ2 variants) and chromosomal abnormalities in loci containing γ-aminobutyric acid receptor (GABAR) genes (e.g. 15q11-13) have been associated with neurodevelopmental phenotypes (Table S1) (Cook et al., 1998). Imbalances in GABAergic and glutamatergic signaling are postulated to contribute to the etiology of neurodevelopmental disorders (reviewed by Sohal and Rubenstein, 2019), and a recent study identifying genes with altered expression trajectories in ASD further hints at crucial roles for ion channels and GABAergic neurons in ASD pathophysiology (Berto et al., 2022).

In addition to ion channel mutations, activity-dependent transcriptional regulation, which is dependent on calcium, has been implicated in neurodevelopmental psychiatric disorders (Boulting et al., 2021; Sanchez-Priego et al., 2022). Altered activity-dependent splicing networks have also been reported in ASD cohorts (Gandal et al., 2018; Parikshak et al., 2016; Quesnel-Vallières et al., 2016). Finally, even in examples where known calcium signaling effectors are not mutated, alterations in calcium handling have been observed in cells from individuals with neurodevelopmental conditions. For example, cortical organoids from patients with 22q11.2 deletion syndrome, a highly penetrant cause of neuropsychiatric disease, exhibit calcium signaling deficits (Khan et al., 2020). Transplanted human induced pluripotent stem cell (iPSC)-derived astrocytes from ASD individuals also display elevated calcium responses (Allen et al., 2022). These observations suggest that calcium signaling may be a convergence point for multiple developmental neuropsychiatric disorders.

Development of the cerebral cortex involves a series of spatiotemporally regulated cellular events, including neural stem and progenitor cell (NSPC) proliferation (reviewed by Libé-Philippot and Vanderhaeghen, 2021; Lin et al., 2021; Llorca and Marín, 2021), migration of newborn neuroblasts into the cortical plate (CP) (reviewed by Francis and Cappello, 2021; Silva et al., 2019) and the differentiation of these cells into mature, synaptically active neurons and glia (reviewed by Bonnefont and Vanderhaeghen, 2021; Taverna et al., 2014) (Box 2). Spontaneous and agonist-induced calcium elevations, neurotransmitter- and depolarization-evoked calcium influx, and SOCE have been observed at various stages of embryonic and adult NSPC lineage progression (reviewed by Toth et al., 2016; Uhlén et al., 2015). These different forms of calcium entry result in the induction of calcium-dependent transcriptional cascades (e.g. CREB-, MEF2-, NFAT- and NPAS4-dependent gene expression) that contribute to developmental regulation of cellular behaviors (reviewed by Greer and Greenberg, 2008). Achieving a granular picture of how cell type-specific calcium signaling impinges on lineage progression in the developing cortex is essential for understanding development and neurodevelopmental disease. Below, we link calcium responses in developing cortical cell populations with the cellular machinery mediating these signals and controlling different cellular behaviors.

Box 2. Embryonic corticogenesis in rodents and humans

During early development, neuroepithelial cells give rise to radial glial cells (RGCs), proliferative neural stem cells that populate the cortex with neurons and astrocytes (reviewed by Libé-Philippot and Vanderhaeghen, 2021; Lin et al., 2021). RGCs initially divide symmetrically and subsequently switch to asymmetric divisions to generate postmitotic migratory neuroblasts and intermediate progenitor cells (IPCs; Haubensak et al., 2004; Noctor et al., 2004). Residing in the pseudostratified ventricular zone (VZ) adjacent to the ventricles, RGCs maintain contact with the overlying pia through a long radial fiber. Newborn neuroblasts exit the VZ and migrate along RGC fibers to reach their final laminar position. Newly generated IPCs detach from the ventricular surface and migrate into the subventricular zone (SVZ), where they divide symmetrically to produce daughter neurons. Young neuroblasts sequentially exit the VZ/SVZ to build the cortex in an inside-out fashion, terminally differentiating in the cortical plate (CP) (reviewed by Bonnefont and Vanderhaeghen, 2021). Early-generated subplate (SP) and Cajal-Retzius (CR) neurons, residing beneath the CP and in the marginal zone (MZ), respectively, are central to the development of cortical circuits (reviewed by Hoerder-Suabedissen and Molnár, 2015; Kanold, 2019; López-Bendito, 2018). In particular, the transient SP population plays an indispensable role in guiding thalamocortical axons innervating the developing cortex (reviewed by López-Bendito, 2018). Cortical interneurons are generated in the ventral telencephalon, tangentially migrating from their germinal centers into the cortex (reviewed by Lim et al., 2018; Silva et al., 2019). In humans, an expanded germinal zone overlying the VZ/SVZ (the outer SVZ; oSVZ) harbors outer radial glia (oRG), which generate cortical neurons and are thought to contribute to the evolutionary expansion of the human neocortex (Fietz et al., 2010; Hansen et al., 2010). Not depicted are less abundant cell types with important roles in cortical development, including microglia, endothelial cells and pericytes. iSVZ, inner SVZ; IZ, intermediate zone.

Cortical NSPC proliferation

Spontaneous calcium elevations

The proliferative NSPC compartment comprises radial glial cells (RGCs) and intermediate progenitor cells (IPCs) (Box 2), which give rise to cortical excitatory neurons and astrocytes. Residing adjacent to the ventricles, these cells are exposed to environmental stimuli impinging on calcium signaling during development, including growth factors and electrical activity (reviewed by Dehay and Kennedy, 2007; Fame and Lehtinen, 2020). Cortical NSPCs express ion channels, pumps and receptors that generate unique calcium dynamics to contribute to developmental cellular behaviors (Fig. 2). Different patterns of spontaneous calcium elevations are observed in the ventricular zone (VZ): slow rises are contained to individual cells, whereas coordinated transients (calcium waves) propagate across gap junction-coupled, mitotically-active RGCs (Bittman et al., 1997; Lo Turco and Kriegstein, 1991; Owens et al., 2000; Weissman et al., 2004).

Fig. 2.

Calcium-dependent regulation of proliferation during corticogenesis. Calcium imaging and electrophysiological recordings performed on embryonic rodent cortical slice cultures demonstrate that radial glial cells (RGCs) in the ventricular zone (VZ) exhibit spontaneous and induced calcium rises. Purinergic signaling through metabotropic P2Y1 receptors initiates calcium transients that propagate across VZ cells. These calcium waves, which modulate proliferation, require gap junctions and IP3-mediated calcium release. RGC primary cilia protrude into the ventricles, where they are exposed to diffusible growth factors in the CSF that initiate calcium rises and also influence RGC division. Finally, depolarization mediated by GABA and glutamate acting on GABARs and AMPARs, respectively, controls proliferation by inducing calcium rises through VGCCs.

Fig. 2.

Calcium-dependent regulation of proliferation during corticogenesis. Calcium imaging and electrophysiological recordings performed on embryonic rodent cortical slice cultures demonstrate that radial glial cells (RGCs) in the ventricular zone (VZ) exhibit spontaneous and induced calcium rises. Purinergic signaling through metabotropic P2Y1 receptors initiates calcium transients that propagate across VZ cells. These calcium waves, which modulate proliferation, require gap junctions and IP3-mediated calcium release. RGC primary cilia protrude into the ventricles, where they are exposed to diffusible growth factors in the CSF that initiate calcium rises and also influence RGC division. Finally, depolarization mediated by GABA and glutamate acting on GABARs and AMPARs, respectively, controls proliferation by inducing calcium rises through VGCCs.

Spontaneous calcium rises are dependent on internal calcium stores, as they persist in the absence of extracellular calcium but are eliminated upon ER calcium depletion (Owens and Kriegstein, 1998). VGCC activation, neurotransmitter signaling and depolarization are not necessary to promote spontaneous rises (Owens and Kriegstein, 1998; Weissman et al., 2004). Instead, initiation of these calcium transients requires purinergic signaling via metabotropic P2Y1 ATP receptors (P2Y1Rs) (Liu et al., 2008; Malmersjö et al., 2013; Owens and Kriegstein, 1998; Owens et al., 2000; Weissman et al., 2004), and NSPCs have been identified as a source of ATP eliciting pro-proliferative calcium responses (Lin et al., 2007). Calcium waves are activated by extracellular ATP in a temporally regulated fashion, occurring robustly at the peak of neurogenesis and propagating across dynamically coupled RGCs via connexin hemichannels at specific cell cycle stages (Bittman et al., 1997; Owens and Kriegstein, 1998; Weissman et al., 2004). Abrogating these waves by antagonizing P2Y1Rs or inhibiting gap junctions significantly reduces proliferation and promotes differentiation (Lin et al., 2007; Malmersjö et al., 2013; Weissman et al., 2004). Single cell RNA-sequencing (scRNA-seq) and calcium imaging studies demonstrate that P2Y1Rs are highly expressed in ventricular RGCs and IPCs of the rodent and human fetal cortex, and that P2Y1R agonists induce calcium rises in these cells (Mayer et al., 2019). P2Y1Rs are downregulated in neurons and, intriguingly, in human outer radial glia (oRG), a neural stem cell population abundant in humans that may contribute to evolutionary expansion of the neocortex, pointing to a conserved role for ATP-dependent calcium signaling in ventricular RGCs and IPCs (Liu et al., 2008; Mayer et al., 2019).

Links between calcium and growth factor signaling

Growth factors are important cell cycle regulators (reviewed by Dehay and Kennedy, 2007) that can induce calcium elevations in proliferative NSPCs. High concentrations of basic fibroblast growth factor (bFGF), for example, elicit robust cytoplasmic calcium rises in the apical end foot and cell body of RGCs, propagating in a sustained manner through the RGC fiber (Rash et al., 2016). bFGF can promote proliferation in concert with epidermal growth factor (EGF) (Tropepe et al., 1999), which stimulates ER calcium store depletion to activate SOCE and induce NFAT-dependent transcription in ganglionic eminence (GE)-derived ventral progenitors and adult subventricular zone (SVZ) cells (Somasundaram et al., 2014). NFATs have been linked to cell cycle regulation (Carvalho et al., 2007; Teixeira et al., 2016) and progenitor proliferation in the neural tube and postnatal neural cultures (Huang et al., 2011; Serrano-Pérez et al., 2015). In GE progenitors, inhibiting SOCE using genetic and pharmacological approaches decreases proliferation (Somasundaram et al., 2014). Although the function and upstream activators of SOCE in cortical NSPCs remain unknown, spontaneous calcium waves in the VZ are partially mediated by intracellular calcium stores and IP3-dependent calcium release (Weissman et al., 2004). Application of the IP3 receptor antagonist 2-aminoethoxydiphenyl borate, at doses that inhibit SOCE, reduces the amplitude and duration of calcium transients in cortical RGCs (Rash et al., 2016), pointing to potential roles for SOCE in proliferative NSPCs.

Neurotransmitter signaling and calcium

Neurotransmitter signaling also influences cortical NSPC calcium elevations to regulate cell division. In the embryonic brain, upon binding to GABAARs, the neurotransmitter GABA depolarizes immature cells (reviewed by Ben-Ari et al., 2007). GABAAR-dependent depolarization results from unopposed developmental activity of the Na+-K+-2Cl transporter NKCC1 (also known as SLC12A2), which yields elevated chloride concentrations in embryonic neural cells (Owens et al., 1996, 1999; reviewed by Ben-Ari et al., 2007). The postnatal emergence of the K+-Cl co-transporter KCC2 (SLC12A5) induces a developmental switch in GABA activity from depolarizing to hyperpolarizing (reviewed by Ben-Ari et al., 2007). This maturation of inhibition is regulated by the activity of the largely transient subplate (SP) neuron population (Kanold and Shatz, 2006), by GABA (Ganguly et al., 2001) and by growth factor signaling (Rivera et al., 2004). The role of neural activity in developmental regulation of intracellular chloride and the transition to GABA inhibition is supported by studies demonstrating that manipulating excitatory input or sensory experience modulates KCC2 expression (He et al., 2010; Kanold and Shatz, 2006; Sernagor et al., 2003). Activity and cytosolic calcium also alter chloride gradients in immature hippocampal neurons (Fiumelli et al., 2005), whereas activity-dependent neurotrophins modulate KCC2 expression and GABAergic inhibition (Aguado et al., 2003; Ludwig et al., 2011).

Embryonic cortical NSPCs express functional GABAA receptors (Lo Turco et al., 1995; Mayer et al., 2019; Owens et al., 1996) and GABA-induced depolarization elicits calcium transients in NSPCs through VGCC activation (Lo Turco et al., 1995; Mayer et al., 2019; Owens et al., 1996, 1999; Panagiotakos et al., 2019). GABA depolarization inhibits DNA synthesis, decreasing cortical NSPC proliferation (Antonopoulos et al., 1997; Lo Turco et al., 1995). This effect can be rescued by application of chloride transport blockers, suggesting that the depolarizing activity of GABA underlies its ability to suppress NSPC proliferation (Lo Turco et al., 1995). Importantly, separately dissecting the effects of GABA in the VZ and SVZ reveals that GABA inhibits SVZ progenitor proliferation but has pro-proliferative effects in the VZ, shortening the cell cycle to promote mitotic re-entry (Haydar et al., 2000). This suggests that GABA exerts cell type-specific effects on RGCs and IPCs, although how they are transduced to influence calcium in each cell type remains unknown. Understanding how developmental GABAergic activity is linked to the electrical properties of progenitor types, which can change across developmental time (Vitali et al., 2018), represents an exciting avenue for future studies.

Likewise, the excitatory neurotransmitter glutamate depolarizes NSPCs, promoting calcium rises that control proliferation. Although NMDARs are expressed in the developing brain (Henson et al., 2008; Lo Turco et al., 1991; Monyer et al., 1994), a confluence of data points to NMDARs playing an outsized role in post-mitotic neurons compared with NSPCs (Behar et al., 1999; Lo Turco et al., 1991, 1995; Maric et al., 2000; Mayer et al., 2019). In the rat VZ, NMDA does not elicit currents in NSPCs, whereas in human RGCs and oRG, NMDA elicits magnesium-insensitive currents at resting membrane potentials (Lo Turco et al., 1991, 1995; Maric et al., 2000; Mayer et al., 2019). Calcium imaging of human fetal cells identified a small fraction of VZ cells responding to NMDA, but scRNA-seq reveals that these cells are predominantly excitatory neurons and mostly absent from NSPC clusters (Mayer et al., 2019).

In contrast, ionotropic α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) and kainate (KA) receptors are expressed in VZ/SVZ cells (Haydar et al., 2000; Lo Turco et al., 1995; Maric et al., 2000). Four main subunits, GluA1-4, compose heteromeric AMPA receptors (AMPARs) (reviewed by Traynelis et al., 2010), and the presence of GluA2 renders AMPARs calcium impermeable (Sommer et al., 1991). Intriguingly, GluA2 abundance increases postnatally, suggesting that embryonic neural cells are more likely to express calcium-permeable AMPARs (Kumar et al., 2002; reviewed by Behm and Öhman, 2016). In line with this, AMPA induces calcium elevations in isolated NSPCs completing their final division (Maric et al., 2000), and AMPA/KA antagonists block calcium elevations elicited by glutamate in rat cortical VZ cells (Lo Turco et al., 1995). AMPAR stimulation in isolated human fetal cortical NSPCs also promotes differentiation (Mayer et al., 2019; Whitney et al., 2008). Consistent with these findings, glutamate and KA (but not NMDA) significantly inhibit DNA synthesis in VZ cells (Lo Turco et al., 1995). Once again, separating RGCs from IPCs reveals a more complex picture: glutamate increases proliferation in the VZ and decreases SVZ proliferation, highlighting differential responses of distinct progenitor populations to neurotransmitter signaling (Haydar et al., 2000). It will be important to dissect these responses across development, particularly in light of data demonstrating progressive RGC hyperpolarization during cortical neurogenesis as a key regulator of RGC output (Vitali et al., 2018). Notably, among the channels upregulated to promote this developmental change in RGC membrane potential are several calcium-activated K+ channels (Vitali et al., 2018). Although dynamic changes in K+ channel expression and membrane potential have been linked to the control of proliferation (reviewed by Blackiston et al., 2009), how such changes influence NSPC calcium signaling remains unclear.

The neurotransmitters serotonin (5-HT) and acetylcholine (Ach) also induce NSPC calcium transients. Maternally-derived 5-HT is reported to regulate proliferation in the developing brain (Côté et al., 2007); however, stimulation of the 5-HT receptors HTR2A and HTR2C promotes rat cortical progenitor survival without affecting proliferation (Dooley et al., 1997). HTR2A is highly expressed in human cortical germinal zones, and the HTR2A agonist TCB-2 induces robust calcium rises in human NSPCs (Mayer et al., 2019). HTR2A inhibition alters fiber length in proliferating human RGCs but does not impact division, positing a human-specific role for serotonergic signaling in maintaining RGC structural integrity during proliferation (Mayer et al., 2019). In contrast, muscarine and Ach stimulate calcium influx in cortical NSPCs via muscarinic Ach receptor (mAchR) activation, and mAchR antagonists and calcium chelators attenuate NSPC proliferation (Atluri et al., 2001; Ma et al., 2000). In GE-derived progenitors, Ach, like EGF, stimulates SOCE via mAchRs, and abrogating SOCE reduces proliferation, pointing to potential links between cholinergic activity, SOCE and NSPC divisions (Somasundaram et al., 2014).

It is possible that agonist-induced SOCE and depolarization-induced calcium entry represent mechanisms that antagonistically regulate NSPC proliferation. Understanding the coordination of different modes of calcium entry and how their interplay impacts proliferative NSPCs is thus essential. Moreover, as calcium influx via VGCCs and SOCE can contribute to calcium oscillations, it will be crucial to determine how cellular responses are encoded in patterns of calcium transients and how these dynamics enable signaling specificity in NSPCs.

Calcium-dependent cellular motility

NSPC motility

Proliferative RGCs undergo interkinetic nuclear migration (INM) – dynamic somatic movements in phase with the cell cycle (reviewed by Taverna et al., 2014). During G1, the somata of RGCs move away from the ventricle to complete S-phase, whereas they move apically during G2, initiating mitosis at the ventricular wall. Pharmacological inhibition of connexin hemichannels suppresses RGC calcium waves (Liu et al., 2010; Weissman et al., 2004) and significantly attenuates INM (Liu et al., 2010). Chelating intracellular calcium also reduces INM distance and speed (Liu et al., 2010), suggesting that calcium propagating through coupled RGCs plays a role in the dynamic changes associated with RGC motility.

IPCs do not exhibit INM, instead delaminating from the VZ and moving into the SVZ along RGC fibers (Noctor et al., 2004; reviewed by Taverna et al., 2014). Pharmacological inhibition of purinergic signaling or P2Y receptor knockdown reduces calcium transient frequency in proliferative IPCs, preventing their migration into the SVZ (Liu et al., 2008). Thus, ATP-mediated calcium signaling is not only necessary for NSPC proliferation, but also for the dynamic progenitor movements that shape the developing cortical cytoarchitecture (Liu et al., 2008).

Spontaneous calcium rises in postmitotic migratory cells

Spontaneous calcium rises have been linked to the radial migration of excitatory neuroblasts (Box 2; Fig. 3). Newborn neuroblasts exiting the VZ exhibit the highest frequency of somatic bursting calcium transients in the developing cortex (Rash et al., 2016). It has also been postulated that calcium transients in RGC fibers, which act as a scaffold for radial migration (Box 2), signal to neuroblasts to influence their calcium dynamics and migratory behavior (Rash et al., 2016). It remains unclear, however, how calcium dynamics in RGCs and neuroblasts intersect, and what the functional contribution of bursting transients in migratory neuroblasts is to radial migration.

Fig. 3.

Calcium and excitatory neuroblast migration. Excitatory neuroblasts undergo radial migration along RGC fibers to reach their final laminar destination. Migratory neuroblasts exiting the VZ display high frequency spontaneous calcium transients. As they enter the SVZ, neuroblasts adopt a multipolar morphology and exhibit low amplitude calcium events. Calcium transients during neuroblast migration are largely mediated by extracellular agonists like BDNF, which activates TrkB, by neurotransmitter receptors (e.g. NMDARs, GABARs), and by downstream activation of VGCCs. Glutamate influences radial migration by inducing calcium influx primarily through NMDARs. Depolarization via GABARs and calcium influx via VGCCs also control radial migration. Emerging evidence suggests that calcium transients propagating through RGC fibers may also contribute to the regulation of neuroblast migration into the cortical plate.

Fig. 3.

Calcium and excitatory neuroblast migration. Excitatory neuroblasts undergo radial migration along RGC fibers to reach their final laminar destination. Migratory neuroblasts exiting the VZ display high frequency spontaneous calcium transients. As they enter the SVZ, neuroblasts adopt a multipolar morphology and exhibit low amplitude calcium events. Calcium transients during neuroblast migration are largely mediated by extracellular agonists like BDNF, which activates TrkB, by neurotransmitter receptors (e.g. NMDARs, GABARs), and by downstream activation of VGCCs. Glutamate influences radial migration by inducing calcium influx primarily through NMDARs. Depolarization via GABARs and calcium influx via VGCCs also control radial migration. Emerging evidence suggests that calcium transients propagating through RGC fibers may also contribute to the regulation of neuroblast migration into the cortical plate.

Unlike excitatory neurons, inhibitory cortical interneurons are generated in subpallial structures, including the medial and caudal ganglionic eminences (MGE and CGE, respectively), undertaking saltatory tangential migration to reach the cortex (reviewed by Buchsbaum and Cappello, 2019; Contractor et al., 2021; Lim et al., 2018; Silva et al., 2019). Within the cortex, interneurons undergo both tangential and radial migration to their appropriate laminar destination (Box 2) (Tanaka et al., 2006; reviewed by Silva et al., 2019). Migratory interneurons exhibit spontaneous calcium transients characterized by oscillatory bursts or individual spikes (Bortone and Polleux, 2009; Martini and Valdeolmillos, 2010). Chelating intracellular calcium or pharmacologically blocking VGCC activity significantly disrupts cytoskeletal dynamics and impairs interneuron migration (Bortone and Polleux, 2009; Martini and Valdeolmillos, 2010). Caffeine-induced ER calcium release also promotes cytoskeletal changes to stimulate interneuron motility (Martini and Valdeolmillos, 2010). Interestingly, SOCE is attenuated in migratory neuroblasts isolated from the GE (Somasundaram et al., 2014), again raising the question of how different modes of calcium entry intersect in developing neurons to coordinate migration.

Agonist-induced calcium rises in migratory neuroblasts

Tyrosine receptor kinase B (TrkB; Ntrk2), canonically bound by brain derived neurotrophic factor (BDNF), has been linked to embryonic cortical neuroblast migration (Bartkowska et al., 2007; Behar et al., 1997) (Fig. 3). BDNF stimulates calcium transients and increases cortical neuroblast migration, which can be blocked by application of Trk inhibitors or calcium chelators (Behar et al., 1997). This suggests that TrkB activation promotes migration partly by inducing downstream calcium signaling, although it should be noted that EGF robustly transactivates TrkB in vivo to stimulate cortical neuroblast migration (Puehringer et al., 2013).

TrkB is also involved in tangential interneuron migration (reviewed by Contractor et al., 2021; Silva et al., 2019). BDNF and neurotrophin 4 (NTF4) activate interneuron TrkB receptors, and Trk inhibitor application reduces the fraction of MGE-derived cells migrating into the cortex. In mice lacking Trkb, the number of calbindin-positive interneurons that reach the cortex is attenuated (Polleux et al., 2002). In contrast to TrkB-mediated activation of PLCγ and MAP kinase in excitatory neuroblasts, BDNF activates PI3-kinase signaling in interneurons to control tangential migration (Polleux et al., 2002). Intriguingly, BDNF- and NTF4-dependent TrkB activation has been implicated in KCC2 regulation, positing a link between growth factor signaling and excitability in developing neurons (Rivera et al., 2004).

Neurotransmitter-induced calcium signaling in migrating neuroblasts

Intracellular calcium modulation via GABAR-mediated signaling also impacts radially migrating cortical neurons. In excitatory neuroblasts, GABA induces calcium transients and stimulates migration (Fig. 3) (Behar et al., 1996). BAPTA blocks these effects, suggesting that calcium transduces GABA stimulation into migratory behavior (Behar et al., 1996). Indeed, GABAA and GABAB receptors are functionally expressed in migratory cortical neurons (López-Bendito et al., 2002; Owens et al., 1996, 1999). Pharmacological studies in embryonic rat cortical slices indicate that activation of different GABARs plays distinct region-specific roles across the cerebral wall (Behar et al., 2000). Similar observations have been replicated in vivo, where local application of GABAAR antagonists or chronic administration of desensitizing levels of GABAAR agonists attenuate spontaneous calcium transients, resulting in cortical heterotopias (Heck et al., 2007). Notably, although GABA has been reproducibly implicated in migration, mice lacking Gad65 (Gad2) and Gad67 (Gad1) do not display major cortical malformations (Ji et al., 1999), and radial migration is not significantly altered in Gad67 knockout mice (Furukawa et al., 2014). Other endogenous activators of GABAARs (e.g. taurine) have thus been proposed to control neuronal migration (Furukawa et al., 2014).

Tangentially migrating cortical interneurons also display GABAR-mediated calcium transients (Soria and Valdeolmillos, 2002). As immature interneurons migrate from the MGE, GABA exerts a depolarizing effect via GABAARs (Bortone and Polleux, 2009; Cuzon et al., 2006), and antagonizing GABAARs results in accumulation of migrating interneurons at the pallial-subpallial boundary (Cuzon et al., 2006). GABA depolarization activates VGCCs to promote calcium influx, acting as a pro-migratory signal, presumably via calcium-dependent pathways to the cytoskeleton (Bortone and Polleux, 2009). During the first postnatal week in mice, KCC2 upregulation renders GABA hyperpolarizing, leading to decreased calcium transients and reduced interneuron motility to terminate migration (Bortone and Polleux, 2009). Suppressing the activity of CGE-derived interneurons at distinct developmental stages does not affect tangential migration but reveals essential contributions of activity-dependent signaling in the radial migration of specific subpopulations into their final laminar positions in the CP (De Marco García et al., 2011).

Glycine receptor (GlyR) activation by endogenous ligands (e.g. glycine, taurine) is also depolarizing in cortical neuroblasts, eliciting calcium rises that may modulate migration (Avila et al., 2013; Flint et al., 1998; Yoshida et al., 2004). Although cortices from mice lacking the developmentally enriched α2 GlyR display no gross morphological defects (Young-Pearse et al., 2006), genetic deletion of these receptors results in impaired interneuron migration (Avila et al., 2013). In migratory cortical interneurons, extrasynaptic glycine acting on α2 GlyRs stimulates dynamic calcium fluctuations via VGCCs, promoting motility through calcium-dependent tuning of actomyosin contractions (Avila et al., 2013). In excitatory neuroblasts, the role of GlyRs is less clear (Furukawa et al., 2014), although GlyR activation in organotypic slice cultures, in the presence of glycine uptake inhibitors, impedes radial migration (Nimmervoll et al., 2011).

Seminal experiments in cerebellar granule cells first demonstrated calcium-dependent roles for glutamate in neuroblast migration (Komuro and Rakic, 1993). In the developing cerebral cortex, glutamate released by post-mitotic neurons induces calcium transients in radially migrating neuroblasts, modulating their motility primarily through NMDARs (Fig. 3) (Behar et al., 1999; Hirai et al., 1999). NMDARs are heteromers, the majority consisting of two GluN1 and two GluN2 subunits. Whereas GluN1 subunits are expressed before and after birth in the cortex, GluN2 subunit expression is dynamically regulated during development, resulting in NMDARs with markedly different physiological properties. Compared with those containing GluN2B subunits, GluN2A-containing NMDARs have faster deactivation kinetics, higher open probabilities and lower sensitivity to agonists. Although both NMDAR subtypes display similar calcium permeability, their unique gating properties shape the dynamics of their contribution to calcium influx to influence downstream signaling (Erreger et al., 2005; Sheng et al., 1994; reviewed by Paoletti et al., 2013; Wyllie et al., 2013). In humans, GRIN2B, encoding GluN2B, is highly expressed in postmitotic embryonic neurons, whereas GRIN2A, encoding GluN2A, is expressed in embryonic RGCs and neurons after birth (Mayer et al., 2019). This developmental subunit switch, which occurs in early postnatal life in the rodent and can be regulated by neural activity (reviewed by Yashiro and Philpot, 2008), results in enriched GluN2B abundance during the peak of neuroblast migration. Consequently, shRNA-dependent GluN2B and GluN1 knockdown in embryonic rodent cortices delays neuronal migration, whereas manipulating GluN2A does not affect neuroblast motility (Jiang et al., 2015). Pharmacological NMDAR inhibition or calcium chelators in the presence of NMDA also abrogate neuroblast migration (Behar et al., 1999; Hirai et al., 1999; Reiprich et al., 2005; Yuryev et al., 2018). More recently, transient glutamatergic transmission from SP neurons onto excitatory neuroblasts was shown to regulate neuroblast migration in an NMDAR-dependent manner (Ohtaka-Maruyama et al., 2018). Although these studies suggest that NMDARs are essential for radial migration, genetic inactivation of Grin1, encoding GluN1, reveals no major deficits in neuronal distribution (Messersmith et al., 1997; Iwasato et al., 2000). This may result from mechanisms compensating for long-term GluN1 loss of function (reviewed by Luhmann et al., 2015; Medvedeva and Pierani, 2020). In tangentially migrating cortical interneurons, NMDA and Kainate also induce calcium transients (Soria and Valdeolmillos, 2002), and activation of NMDARs or AMPARs positively regulates interneuron motility in a VGCC-dependent manner (Bortone and Polleux, 2009). Within the intermediate zone (IZ), tangentially migrating interneurons continue to express calcium-permeable AMPARs (Métin et al., 2000) and, in CGE-derived interneuron subtypes, 5-HT3A receptor activation induces calcium transients required for proper migration (Murthy et al., 2014).

VGCCs in neuroblast migration

A function for VGCCs in neuronal migration was initially described in cerebellar granule cells (Komuro and Rakic, 1992). In the embryonic cortex, L-type VGCCs are highly expressed in cortical neurons, and migrating upper layer neurons exhibit spontaneous L-type VGCC-mediated calcium transients (Kamijo et al., 2018). In utero Cav1.2 overexpression in excitatory neurons destined for upper cortical layers impairs radial migration, and a severe calcium influx-dependent migratory defect in this population results from electroporation of Cav1.2 channels bearing the TS mutation (Kamijo et al., 2018).

L-type VGCCs, and to a lesser extent N-type VGCCs, are also essential for interneuron motility (Bortone and Polleux, 2009). Depolarization of immature interneurons by GABA or glutamate activates VGCCs to promote tangential migration into the cortex (Bortone and Polleux, 2009). Assembloid models using human iPSCs further support a role for Cav1.2 in human cortical interneuron migration, demonstrating that inhibitory neurons from TS patients display abnormal migratory behaviors (Birey et al., 2017). Dissecting these migratory phenotypes demonstrates that VGCC-dependent calcium signaling impinges on distinct molecular networks to influence cellular motility (Birey et al., 2022).

Calcium and programmed cell death

Developing cortical circuits are refined by precisely regulated apoptosis, which scales down NSPC, pyramidal cell and cortical interneuron populations. Approximately 12% of cortical pyramidal cells undergo apoptosis in the early postnatal period in rodents (Wong et al., 2018), and a substantial fraction of cortical interneurons also undergo postnatal cell death (Priya et al., 2018; Southwell et al., 2012). In both developing populations, a central regulator of apoptosis is neuronal activity and intracellular calcium signaling.

In vitro studies in the 1990s first implicated activity-dependent calcium signaling in cortical pyramidal neuron survival (Ghosh et al., 1994; Voigt et al., 1997). Stimulation of embryonic cortical cultures with potassium chloride (KCl) enhances cell survival by promoting neurotrophin expression, an effect that can be eliminated by chelating calcium or pharmacologically blocking L-type VGCCs (Ghosh et al., 1994). NMDARs contribute to developmental apoptosis of pyramidal cells during discrete temporal windows, as early postnatal NMDAR inhibition promotes cell death. This effect can be rescued by concurrent VGCC activation, suggesting that VGCCs and NMDARs have overlapping roles in promoting calcium-dependent survival (Ghosh et al., 1994; Heck et al., 2008; Ikonomidou et al., 1999). The contribution of GABA to cortical neuron survival is again linked to its depolarizing or hyperpolarizing activity. Global NKCC1 inactivation reduces developmental cell death of transient layer 1 Cajal-Retzius (CR) neurons (Blanquie et al., 2017a), and hyperpolarization promotes subtype-specific survival of CR neurons (Riva et al., 2019). Inhibiting activity altogether in developing pyramidal cells significantly reduces their survival (Voigt et al., 1997), and regional variations in apoptosis in vivo are partly regulated by corresponding differences in endogenous activity (Blanquie et al., 2017b). In vitro evidence suggests, however, that it is not simply the presence or absence of activity that regulates apoptosis in developing cortical neurons; rather, distinct activity patterns may modulate survival, further supporting the notion that information encoded in the frequency of calcium signals can drive changes in cell behavior (Golbs et al., 2011; Wong Fong Sang et al., 2021). Investigating how ion channel subunit composition and localization contribute to activity patterns in vivo and how these patterns activate specific calcium-dependent signaling pathways in developing cortical neurons to regulate survival is an essential next step.

Notably, the emergence of synchronous calcium transients within populations of cortical pyramidal and hippocampal neurons is positively correlated with survival (Heck et al., 2008; Murase et al., 2011; Voigt et al., 1997). Such synchronous transients in neuronal domains are partially mediated by gap junctions (Kandler and Katz, 1998; Yuste et al., 1992), and pharmacological gap junction inhibition preserves spontaneous asynchronous activity but increases neuronal apoptosis (Heck et al., 2008). In line with this, multiple calcium-dependent transcription factors have been linked to neuronal survival, including MEF2 (Liu et al., 2003; Mao et al., 1999) and NFATc4 (Benedito et al., 2005; Quadrato et al., 2012).

Increased activity also correlates with improved postnatal survival of cortical interneurons (Denaxa et al., 2018; Priya et al., 2018; Wong et al., 2018, 2022). Interneurons that die are less likely to participate in coordinated network events, displaying fewer synchronized calcium fluctuations in vivo compared with cells that survive (Duan et al., 2020; Wong et al., 2018). Accordingly, artificially hyperpolarizing interneurons decreases their survival, whereas increasing their activity promotes survival (Denaxa et al., 2018; Priya et al., 2018). Interestingly, the contribution of activity-dependent signaling to interneuron survival, thought to be mediated at least partly through the calcium-sensitive CaN/NFAT pathway (Priya et al., 2018), is subtype-specific (Denaxa et al., 2018; Priya et al., 2018). The activity of maturing neuronal populations also non-cell autonomously regulates interneuron subtype survival to shape developing circuits (Wong et al., 2018, 2022). Glutamatergic signaling is required for activity-dependent survival of neurogliaform and basket cells, whereas bipolar cells rely on serotonin to modulate activity-dependent survival (Wong et al., 2022). It will be interesting to determine the calcium-dependent pathways that link electrical signals to the survival of specific neuronal subtypes.

Differentiation

Calcium and electrical activity play central roles in regulating aspects of neuronal identity. We restrict our focus here to a brief discussion of calcium functions in early events underlying cortical neuron differentiation, namely the acquisition of fate determinants and initial elaboration of neurites. The involvement of activity-dependent signaling in later events, such as synaptogenesis, circuit integration, maturation and plasticity, have been extensively reviewed elsewhere (Antón-Bolaños et al., 2018; Greer and Greenberg, 2008; Molnár et al., 2020; Pan and Monje, 2020; Rosenberg and Spitzer, 2011).

Pioneering studies in Xenopus spinal neurons demonstrated that intracellular calcium elevations regulate distinct aspects of differentiation, including neurotransmitter specification, maturation of ionic conductances and synaptic development (Borodinsky and Spitzer, 2007; Gu and Spitzer, 1995). In the developing cortex, expression of neuronal fate determinants may be calcium-regulated. Elevated calcium in differentiating human iPSC-derived TS cortical neurons biases neuronal production in vitro (Paşca et al., 2011). Moreover, in vivo gain and loss of function of the L-type VGCC Cav1.2 in differentiating NSPCs bidirectionally modulates the relative abundance of cells expressing markers of callosal or subcerebral projection neurons in a calcium-dependent manner (Panagiotakos et al., 2019). GABAAR-dependent activation of L-type VGCCs also controls the morphological differentiation of pyramidal cells (McAllister et al., 1996; Redmond et al., 2002; Wayman et al., 2006; reviewed by Chen and Ghosh, 2005), and premature KCC2 expression in vivo reveals that GABA depolarization is necessary for dendritic maturation of cortical neurons (Cancedda et al., 2007). In addition, elevating neuronal activity and spontaneous calcium transient frequency in migratory neurons arrests migration and induces precocious dendritic branching (Bando et al., 2016). Consistent with this, thalamocortical afferent activity is indispensable for proper development of neuronal morphology (Callaway and Borrell, 2011; Li et al., 2013), as well as barrel column formation in mice (Antón-Bolaños et al., 2019; Li et al., 2013). Recently, a small molecule chemical screen in human iPSC-derived cortical neurons identified epigenetic modifiers and calcium signaling activators as enhancers of neuronal maturation, including neurite elaboration, further supporting that facilitating calcium-dependent gene expression can promote terminal differentiation of cortical neurons (Ciceri et al., 2022 preprint; Hergenreder et al., 2022 preprint).

Calcium-regulated transcription factors or transcriptional activators, including CREB (Redmond et al., 2002; Wayman et al., 2006), NEUROD (Gaudillière et al., 2004) and CREST (Aizawa et al., 2004), have also been implicated in dendrite development. In addition, a CREB-dependent microRNA (miR132) positively regulates cortical neuron neurite outgrowth (Vo et al., 2005). Callosal axon outgrowth in the developing cortex is impeded by suppressing neuronal activity (Mizuno et al., 2007; Rodríguez-Tornos et al., 2016; Suárez et al., 2014; Wang et al., 2007), and axonal pathfinding has been linked to intracellular calcium signaling involving kinases and phosphatases with distinct calcium sensitivities (reviewed by Gomez and Zheng, 2006). How these calcium-dependent mechanisms cooperate to control axonal and dendritic elaboration in the developing cortex remains unclear. Intriguingly, a calcium-independent interaction between Cav1.2 and RhoA was shown to regulate dendritic morphogenesis (Krey et al., 2013), highlighting additional roles for VGCCs as anchors for large signaling complexes at the membrane.

Electrical activity and neurotransmitter signaling regulate cortical interneuron differentiation in a calcium-dependent manner. BDNF, in conjunction with depolarization, enhances dendritic branching and electrophysiological maturation of parvalbumin (PV)-expressing interneurons (Berghuis et al., 2004). Postnatal electrical activity mediated by ionotropic glutamate receptors is also required for morphological maturation of calretinin- and reelin-expressing cortical interneurons (De Marco García et al., 2011). The activity-regulated DNA binding protein SATB1, which is associated with postnatal survival of somatostatin (SST)-expressing MGE-derived interneurons, is necessary for their maturation and terminal differentiation (Close et al., 2012; Denaxa et al., 2012). KCl-induced SATB1 upregulation is dependent on both calcium influx through L-type VGCCs and GABAR activation (Denaxa et al., 2012). More recently, the calcium-dependent transcription factor MEF2C was found to be required for the differentiation of PV-expressing cortical interneurons (Mayer et al., 2018). Activity-dependent splicing regulators such as the ASD-relevant RNA binding protein RBFOX1 (Table S1), which promote neuronal differentiation of cortical NSPCs (Zhang et al., 2016), also regulate distinct aspects of PV- and SST-expressing interneuron maturation and connectivity (Wamsley et al., 2018). These data reveal that activity-dependent calcium signaling is essential for the acquisition of molecular identity and morphology in developing pyramidal and interneuron populations.

Calcium and gliogenesis

To ensure that neurogenesis proceeds faithfully during early cortical development, RGCs actively inhibit intrinsic mechanisms of astrogliogenesis, including transcription of astrocyte-specific genes (reviewed by Miller and Gauthier, 2007). Extrinsic signals acting via neurotrophic factors later stimulate cortical RGCs to become gliogenic (Qian et al., 1997; Song and Ghosh, 2004), in part via JAK/STAT pathway activation (Bonni et al., 1997). G protein signaling initiated by pituitary adenylate cyclase-activating polypeptide (PACAP, encoded by Adcyap1) and transduced via calcium has been identified as a complementary mechanism involved in the onset of astrogliogenesis (Fig. 4) (Nishimoto et al., 2007; Vallejo and Vallejo, 2002). Activation of PAC1 receptors by PACAP induces cyclic AMP (cAMP) production, which promotes astrocyte differentiation (Cebolla et al., 2008; McManus et al., 1999; Vallejo and Vallejo, 2002). PACAP stimulation elicits gradual intracellular calcium rises in NSPCs, whereas cAMP antagonism eliminates these rises and blocks astrocyte differentiation (Cebolla et al., 2008). During astrogliogenesis, calcium binding to the downstream regulatory element antagonist modulator (DREAM) is required for expression of the astrocyte-specific glial fibrillary acidic protein (Gfap) gene (Cebolla et al., 2008). These data indicate that PACAP induces cAMP-dependent calcium entry, enabling calcium-dependent astrocyte-specific gene transcription (Cebolla et al., 2008; McManus et al., 1999; Vallejo and Vallejo, 2002). PACAP signaling in mice lacking Dream (Kcnip3) fails to induce astrocyte differentiation, but this can be rescued by JAK/STAT activation, suggesting that calcium-regulated DREAM signaling works in parallel to JAK/STAT signaling during astrogliogenesis (Cebolla et al., 2008).

Fig. 4.

Calcium and astrogliogenesis. Although microglia and embryonic oligodendrocytes originate largely outside of the cortex, RGCs (light purple) transition from making neurons to generating cortical astrocytes (green) during mid-to-late gestation in the rodent. This gliogenic switch begins at approximately embryonic day 17 in mice and continues postnatally. Activation of plasma membrane receptors [glycoprotein 130 (Gp130; Il6st), ciliary neurotrophic factor receptor (CNTFR) and leukemia inhibitory factor receptor (LIFR)] regulates Gfap transcription and astroglial differentiation via JAK/STAT signaling. In parallel, pituitary adenylate cyclase activating polypeptide (PACAP)-mediated activation of its receptor, PAC1R, generates cAMP-dependent calcium elevations. Calcium binds DREAM to promote expression of astrocyte-specific genes and astrogliogenesis.

Fig. 4.

Calcium and astrogliogenesis. Although microglia and embryonic oligodendrocytes originate largely outside of the cortex, RGCs (light purple) transition from making neurons to generating cortical astrocytes (green) during mid-to-late gestation in the rodent. This gliogenic switch begins at approximately embryonic day 17 in mice and continues postnatally. Activation of plasma membrane receptors [glycoprotein 130 (Gp130; Il6st), ciliary neurotrophic factor receptor (CNTFR) and leukemia inhibitory factor receptor (LIFR)] regulates Gfap transcription and astroglial differentiation via JAK/STAT signaling. In parallel, pituitary adenylate cyclase activating polypeptide (PACAP)-mediated activation of its receptor, PAC1R, generates cAMP-dependent calcium elevations. Calcium binds DREAM to promote expression of astrocyte-specific genes and astrogliogenesis.

Neural activity and downstream calcium signaling is also implicated in the development and function of cortical oligodendrocyte precursor cells (OPCs) and microglia (Box 3). Dissecting how activity-dependent calcium signaling facilitates communication between developing neurons and glia will enhance our understanding of how the cortex is built.

Box 3. Calcium and embryonic glial cells originating outside the cortex

Embryonic oligodendrocyte precursor cells (OPCs) initially populate the cortex through two waves of migration from the ganglionic eminences (Kessaris et al., 2006). A third group of OPCs is generated in the postnatal cortex, replacing a subset of ventrally-derived OPCs (Kessaris et al., 2006). During these developmental windows, OPCs begin to express ion channels and neurotransmitter receptors that enable them to respond to activity-dependent signals (De Biase et al., 2010; Fulton et al., 2010; Spitzer et al., 2019; reviewed by Bergles and Richardson, 2015). It was first demonstrated in the rodent optic nerve that inhibiting electrical activity reduces the number of mitotic OPCs (Barres and Raff, 1993). Pharmacological, electrophysiological and optogenetic manipulations in the cortex have since reinforced the notion that neuronal activity regulates OPC proliferation and myelination (Demerens et al., 1996; Gary et al., 2012; Gibson et al., 2014; Mitew et al., 2018). The effects of electrical activity on OPCs are likely at least partly transduced via VGCCs, as Cav1.2 loss of function results in impaired OPC proliferation, axon-OPC interactions and myelination (Cheli et al., 2015, 2016). Live imaging of the developing zebrafish spinal cord also reveals that neuronal activity induces different patterns of calcium signals to regulate myelination (Baraban et al., 2017; Krasnow et al., 2018). What initiates these dynamics and how they influence genetic programs driving OPC differentiation and myelination remains unclear. Although OPCs differentially respond to activity-dependent signals in the spinal cord (Marisca et al., 2020), such OPC functional heterogeneity in the developing cortex remains poorly understood.

Microglia originate outside the CNS and migrate into the cortex during embryonic development (Ginhoux et al., 2010; reviewed by Thion and Garel, 2017), where they play important roles in regulating progenitor abundance and sculpting developing circuits (Cunningham et al., 2013; Squarzoni et al., 2014; Ueno et al., 2013; reviewed by Thion and Garel, 2020). Although various neurotransmitters, immune molecules and ligands for purinergic receptors induce microglial calcium rises in vitro (reviewed by Umpierre and Wu, 2021), it remains less clear how calcium influences the functions of cortical microglia. Neuronal activity regulates microglial-mediated synaptic pruning (Schafer et al., 2012), and microglia sensitive to GABA preferentially remodel inhibitory synapses in the postnatal cortex (Favuzzi et al., 2021). In the developing zebrafish spinal cord, activity-dependent myelin phagocytosis by microglia is characterized by spontaneous calcium transients in microglial processes contacting myelin (Hughes and Appel, 2020). Future studies probing how activity and calcium facilitate neuron-glia communication during development will aid our understanding of how different cell types function in concert to build the cortex.

As we have reviewed here, the coordinated spatiotemporal regulation of calcium signaling directs cellular behaviors that underlie early cortical development, including aspects of NSPC function. Calcium is a key mediator of activity-dependent gene expression, serving as a hub linking environmental cues to the cytoskeleton, metabolic pathways and other biochemical cascades. Underscoring these crucial developmental roles, genetic studies reveal that mutations in genes encoding calcium signaling modulators contribute to the pathophysiology of neurodevelopmental disorders. There remain, however, open questions about how activity and calcium-dependent processes are regulated at a cell type-specific level and how this regulation coordinates cellular behaviors during corticogenesis. In other cell and tissue types, properties of intracellular calcium dynamics have been linked to activation of specific downstream transcription factors. It is thus tempting to hypothesize that distinct cell type-specific patterns of electrical signals and calcium elevations in the embryonic cortex may subserve specific developmental roles. A natural corollary to this hypothesis is that disrupting calcium signaling and patterned electrical activity in specific cortical populations during development may promote neurodevelopmental disease.

Much remains to be elucidated about how calcium signaling is initiated and transduced in developing cortical cells. Titration of calcium signaling is not only achieved through dynamic regulation of ion channels and signaling proteins but also via coordination of extrinsic cues, including maternal hormones and metabolic regulation (Rash et al., 2018; Tyzio et al., 2006), which indirectly influence calcium homeostasis. How then do extrinsic signals converge to shape calcium entry in different cortical cell populations? How do different modes of calcium entry work cooperatively to direct gene expression programs across cell types? And how might intracellular calcium signals in one population influence dynamic interactions with other cell types? For example, do compartmentalized calcium signals that propagate across polarized cells, as reported in RGCs, influence local cellular processes (e.g. translation)? Single-cell, long-read and spatial sequencing technologies, organoid modeling, subcellular calcium and voltage imaging approaches, optogenetic and chemogenetic tools, and advances in labeling and isolating developing cortical cell populations will hopefully allow us to address these and other questions with unprecedented cellular resolution.

Determining how disease-relevant mutations affect intracellular signaling and cell type-specific developmental behaviors across space and time (reviewed by Panagiotakos and Pasca, 2022) will also enable the development of therapeutic approaches for neurodevelopmental diseases targeting calcium and activity-dependent mechanisms. Of special interest is determining how early disruption of activity-dependent signaling might cascade into later developmental events to promote disease. Recent findings support the notion that neurodevelopmental mechanisms may be reactivated to promote adult and aging-related disease states (Boxes 4 and 5), reinforcing the significance of exploring how calcium signals are normally regulated to coordinate the development of functional circuits. Looking ahead, it will be important to consider how changes at the organismal level (e.g. immune function, metabolism, gut-brain axis) may contribute to the regulation and misregulation of calcium in the embryonic and adult cortex to better understand normal development and the emergence of neurological disorders.

Box 4. Reactivation of developmental calcium signaling mechanisms in glioma

In primary glioma specimens from adult human patients, stem-like glioma cells display molecular signatures reflecting neural stem and progenitor cell (NSPC) identity (Bhaduri et al., 2020; Venteicher et al., 2017; Wang et al., 2020), and their morphology, behavior and lineage trajectory resembles that of embryonic radial glial cells (RGCs) (Bhaduri et al., 2020; Couturier et al., 2020; Wang et al., 2020). Similarities between cortical RGCs and stem-like glioma cells extend into calcium signaling dynamics, suggesting that calcium-regulated developmental mechanisms may be reused to promote glioma initiation and maintenance. For example, patient-derived glioblastoma xenografts in mice display synchronous calcium transients (Venkataramani et al., 2019; Venkatesh et al., 2019) and, as in cortical radial glia, calcium and electrical activity modulates glioma cell proliferation (Urso et al., 2019; Venkatesh et al., 2019; Zhang et al., 2012). Adjacent glioma cells are connected via ‘microtubes’ composed of gap junctions (Osswald et al., 2015), and calcium propagates across coupled cells in a manner reminiscent of calcium waves in the developing cortical ventricular zone (Osswald et al., 2015; Venkataramani et al., 2019). It is plausible that unraveling the calcium signaling mechanisms directing embryonic NSPC proliferation and migration will provide crucial insights into glioma cell biology to identify potential therapeutic targets for the management of glioma invasion and progression.

Box 5. Implications for neurodegenerative diseases

Calcium signaling deregulation has been implicated in neurodegenerative disorders, including Alzheimer's disease (AD) (reviewed by Pchitskaya et al., 2018; Verkhratsky, 2019). In human induced pluripotent stem cell models of AD and frontotemporal lobar degeneration-tauopathy, activity-dependent calcium elevations are abnormally high (Imamura et al., 2016; Park et al., 2018). Neurons and glia also display elevated basal calcium in AD mouse models (reviewed by Pchitskaya et al., 2018; Verkhratsky, 2019), and genes related to increased cytosolic calcium are enriched in patients with heightened risk for sporadic AD (Heck et al., 2015). Intriguingly, calcium- and activity-dependent pathways altered in neurodevelopmental diseases are thought to be affected in AD (Ivashko-Pachima et al., 2021; Mencer et al., 2021). Key calcium signaling effectors like DYRK1A (Table S1) are deregulated both in neurodevelopmental disorders and cortices of sporadic AD patients (Ferrer et al., 2005). Consistent with this, aging and AD rodent models exhibit aberrant calcium-dependent CaN/NFAT signaling (Foster et al., 2001; Norris et al., 2005), and blocking CaN/NFAT in AD models improves synaptic function, amyloid pathology and astrogliosis (Furman et al., 2012).

Emerging studies also suggest that individuals with neurodevelopmental disorders such as autism spectrum disorder (ASD) and Down syndrome (DS) have increased risk of developing AD and related dementias early in life (Vivanti et al., 2021; reviewed by Lott and Head, 2019). Although, in the case of DS, association with early-onset dementia is related to increased amyloid precursor protein (APP) resulting from trisomy of all or part of chromosome 21, multiple genetic and environmental factors likely contribute to increased AD susceptibility. Deregulation of electrical activity, which impinges on calcium, represents one possible contributing mechanism common to neurodevelopmental and neurodegenerative disorders. In DS and ASD mouse models, abnormalities in NKCC1/KCC2 expression promote persistent depolarizing activity of GABA, akin to the developing brain, in adult and postnatal hippocampal neurons (Deidda et al., 2015; Tyzio et al., 2014). Consequently, administration of the NKCC1 antagonist bumetanide in these models restores chloride gradients and rescues cognitive and behavioral abnormalities (Deidda et al., 2015; Tyzio et al., 2014). Intriguingly, bumetanide was recently shown to improve pathological and behavioral deficits in an AD rodent model (Taubes et al., 2021). Furthermore, APP has been linked to the regulation of KCC2, GABAR and VGCC expression (Chen et al., 2017; Doshina et al., 2017; Yang et al., 2009), implicating it as a regulator of electrical activity and calcium homeostasis.

We thank all members of the Panagiotakos lab past and present for critical discussions pertaining to calcium signals in the developing brain. We also thank T. Palmer and I. Graef for providing constructive feedback on our manuscript. We apologize to any colleagues whose work we were unable to cite due to space constraints.

Funding

Our research is supported by the Sandler Foundation (University of California, San Francisco Sandler Faculty Fellows Program, to G.P.) and the National Institutes of Health (R01 MH125004 and R56/R01 MH127075 to G.P.). Open Access funding provided by the Sandler Foundation and the National Institutes of Health (NIH R01 MH125004). Deposited in PMC for immediate release.

Abernethy
,
D. R.
and
Soldatov
,
N. M.
(
2002
).
Structure-functional diversity of human L-type Ca2+ channel: perspectives for new pharmacological targets
.
J. Pharmacol. Exp. Ther.
300
,
724
-
728
.
Aguado
,
F.
,
Carmona
,
M. A.
,
Pozas
,
E.
,
Aguiló
,
A.
,
Martínez-Guijarro
,
F. J.
,
Alcantara
,
S.
,
Borrell
,
V.
,
Yuste
,
R.
,
Ibañez
,
C. F.
and
Soriano
,
E.
(
2003
).
BDNF regulates spontaneous correlated activity at early developmental stages by increasing synaptogenesis and expression of the K+/Cl- co-transporter KCC2
.
Development
130
,
1267
-
1280
.
Aizawa
,
H.
,
Hu
,
S.-C.
,
Bobb
,
K.
,
Balakrishnan
,
K.
,
Ince
,
G.
,
Gurevich
,
I.
,
Cowan
,
M.
and
Ghosh
,
A.
(
2004
).
Dendrite development regulated by CREST, a calcium-regulated transcriptional activator
.
Science
303
,
197
-
202
.
Allen
,
M.
,
Huang
,
B. S.
,
Notaras
,
M. J.
,
Lodhi
,
A.
,
Barrio-Alonso
,
E.
,
Lituma
,
P. J.
,
Wolujewicz
,
P.
,
Witztum
,
J.
,
Longo
,
F.
,
Chen
,
M.
et al. 
(
2022
).
Astrocytes derived from ASD individuals alter behavior and destabilize neuronal activity through aberrant Ca2+ signaling
.
Mol. Psychiatry
27
,
2470
-
2484
.
An
,
P.
and
Grabowski
,
P. J.
(
2007
).
Exon silencing by UAGG motifs in response to neuronal excitation
.
PLoS Biol.
5
,
e36
.
Antón-Bolaños
,
N.
,
Espinosa
,
A.
and
López-Bendito
,
G.
(
2018
).
Developmental interactions between thalamus and cortex: a true love reciprocal story
.
Curr. Opin. Neurobiol.
52
,
33
-
41
.
Antón-Bolaños
,
N.
,
Sempere-Ferràndez
,
A.
,
Guillamón-Vivancos
,
T.
,
Martini
,
F. J.
,
Pérez-Saiz
,
L.
,
Gezelius
,
H.
,
Filipchuk
,
A.
,
Valdeolmillos
,
M.
and
López-Bendito
,
G.
(
2019
).
Prenatal activity from thalamic neurons governs the emergence of functional cortical maps in mice
.
Science
364
,
987
-
990
.
Antonopoulos
,
J.
,
Pappas
,
I. S.
and
Parnavelas
,
J. G.
(
1997
).
Activation of the GABAA receptor inhibits the proliferative effects of bFGF in cortical progenitor cells
.
Eur. J. Neurosci.
9
,
291
-
298
.
Atluri
,
P.
,
Fleck
,
M. W.
,
Shen
,
Q.
,
Mah
,
S. J.
,
Stadfelt
,
D.
,
Barnes
,
W.
,
Goderie
,
S. K.
,
Temple
,
S.
and
Schneider
,
A. S.
(
2001
).
Functional nicotinic acetylcholine receptor expression in stem and progenitor cells of the early embryonic mouse cerebral cortex
.
Dev. Biol
240
,
143
-
156
.
Avila
,
A.
,
Vidal
,
P. M.
,
Dear
,
T. N.
,
Harvey
,
R. J.
,
Rigo
,
J.-M.
and
Nguyen
,
L.
(
2013
).
Glycine receptor α2 subunit activation promotes cortical interneuron migration
.
Cell Rep.
4
,
738
-
750
.
Bading
,
H.
,
Ginty
,
D. D.
and
Greenberg
,
M. E.
(
1993
).
Regulation of gene expression in hippocampal neurons by distinct calcium signaling pathways
.
Science
260
,
181
-
186
.
Bakowski
,
D.
and
Parekh
,
A. B.
(
2000
).
Voltage-dependent conductance changes in the store-operated Ca2+ current ICRAC in rat basophilic leukaemia cells
.
J. Physiol.
529
,
295
-
306
.
Bando
,
Y.
,
Irie
,
K.
,
Shimomura
,
T.
,
Umeshima
,
H.
,
Kushida
,
Y.
,
Kengaku
,
M.
,
Fujiyoshi
,
Y.
,
Hirano
,
T.
and
Tagawa
,
Y.
(
2016
).
Control of spontaneous Ca2+ transients is critical for neuronal maturation in the developing neocortex
.
Cereb. Cortex
26
,
106
-
117
.
Baraban
,
M.
,
Koudelka
,
S.
and
Lyons
,
D. A.
(
2017
).
Ca2+ activity signatures of myelin sheath formation and growth in vivo
.
Nat. Neurosci.
21
,
19
-
23
.
Barres
,
B. A.
and
Raff
,
M. C.
(
1993
).
Proliferation of oligodendrocyte precursor cells depends on electrical activity in axons
.
Nature
361
,
258
-
260
.
Bartkowska
,
K.
,
Paquin
,
A.
,
Gauthier
,
A. S.
,
Kaplan
,
D. R.
and
Miller
,
F. D.
(
2007
).
Trk signaling regulates neural precursor cell proliferation and differentiation during cortical development
.
Development
134
,
4369
-
4380
.
Behar
,
T. N.
,
Li
,
Y. X.
,
Tran
,
H. T.
,
Ma
,
W.
,
Dunlap
,
V.
,
Scott
,
C.
and
Barker
,
J. L.
(
1996
).
GABA stimulates chemotaxis and chemokinesis of embryonic cortical neurons via calcium-dependent mechanisms
.
J. Neurosci.
16
,
1808
-
1818
.
Behar
,
T. N.
,
Dugich-Djordjevic
,
M. M.
,
Li
,
Y. X.
,
Ma
,
W.
,
Somogyi
,
R.
,
Wen
,
X.
,
Brown
,
E.
,
Scott
,
C.
,
McKay
,
R. D.
and
Barker
,
J. L.
(
1997
).
Neurotrophins stimulate chemotaxis of embryonic cortical neurons
.
Eur. J. Neurosci.
9
,
2561
-
2570
.
Behar
,
T. N.
,
Scott
,
C. A.
,
Greene
,
C. L.
,
Wen
,
X.
,
Smith
,
S. V.
,
Maric
,
D.
,
Liu
,
Q.-Y.
,
Colton
,
C. A.
and
Barker
,
J. L.
(
1999
).
Glutamate acting at NMDA receptors stimulates embryonic cortical neuronal migration
.
J. Neurosci.
19
,
4449
-
4461
.
Behar
,
T. N.
,
Schaffner
,
A. E.
,
Scott
,
C. A.
,
Greene
,
C. L.
and
Barker
,
J. L.
(
2000
).
GABA receptor antagonists modulate postmitotic cell migration in slice cultures of embryonic rat cortex
.
Cereb. Cortex
10
,
899
-
909
.
Behm
,
M.
and
Öhman
,
M.
(
2016
).
RNA Editing: a contributor to neuronal dynamics in the mammalian brain
.
Trends Genet.
32
,
165
-
175
.
Ben-Ari
,
Y.
,
Gaiarsa
,
J.-L.
,
Tyzio
,
R.
and
Khazipov
,
R.
(
2007
).
GABA: a pioneer transmitter that excites immature neurons and generates primitive oscillations
.
Physiol. Rev.
87
,
1215
-
1284
.
Benedito
,
A. B.
,
Lehtinen
,
M.
,
Massol
,
R.
,
Lopes
,
U. G.
,
Kirchhausen
,
T.
,
Rao
,
A.
and
Bonni
,
A.
(
2005
).
The transcription factor NFAT3 mediates neuronal survival
.
J. Biol. Chem.
280
,
2818
-
2825
.
Berghuis
,
P.
,
Dobszay
,
M. B.
,
Sousa
,
K. M.
,
Schulte
,
G.
,
Mager
,
P. P.
,
Härtig
,
W.
,
Görcs
,
T. J.
,
Zilberter
,
Y.
,
Ernfors
,
P.
and
Harkany
,
T.
(
2004
).
Brain-derived neurotrophic factor controls functional differentiation and microcircuit formation of selectively isolated fast-spiking GABAergic interneurons
.
Eur. J. Neurosci.
20
,
1290
-
1306
.
Bergles
,
D. E.
and
Richardson
,
W. D.
(
2015
).
Oligodendrocyte Development and Plasticity
.
Cold Spring Harb. Perspect. Biol.
8
,
a020453
.
Berridge
,
M. J.
,
Bootman
,
M. D.
and
Roderick
,
H. L.
(
2003
).
Calcium signalling: dynamics, homeostasis and remodelling
.
Nat. Rev. Mol. Cell Biol.
4
,
517
-
529
.
Berto
,
S.
,
Treacher
,
A. H.
,
Caglayan
,
E.
,
Luo
,
D.
,
Haney
,
J. R.
,
Gandal
,
M. J.
,
Geschwind
,
D. H.
,
Montillo
,
A. A.
and
Konopka
,
G.
(
2022
).
Association between resting-state functional brain connectivity and gene expression is altered in autism spectrum disorder
.
Nat. Commun.
13
,
3328
.
Bhaduri
,
A.
,
Di Lullo
,
E.
,
Jung
,
D.
,
Müller
,
S.
,
Crouch
,
E. E.
,
Espinosa
,
C. S.
,
Ozawa
,
T.
,
Alvarado
,
B.
,
Spatazza
,
J.
,
Cadwell
,
C. R.
et al. 
(
2020
).
Outer radial glia-like cancer stem cells contribute to heterogeneity of glioblastoma
.
Cell Stem Cell
26
,
48
-
63.e6
.
Bhat
,
S.
,
Dao
,
D. T.
,
Terrillion
,
C. E.
,
Arad
,
M.
,
Smith
,
R. J.
,
Soldatov
,
N. M.
and
Gould
,
T. D.
(
2012
).
CACNA1C (Cav1.2) in the pathophysiology of psychiatric disease
.
Prog. Neurobiol.
99
,
1
-
14
.
Birey
,
F.
,
Andersen
,
J.
,
Makinson
,
C. D.
,
Islam
,
S.
,
Wei
,
W.
,
Huber
,
N.
,
Fan
,
H. C.
,
Metzler
,
K. R. C.
,
Panagiotakos
,
G.
,
Thom
,
N.
et al. 
(
2017
).
Assembly of functionally integrated human forebrain spheroids
.
Nature
545
,
54
-
59
.
Birey
,
F.
,
Li
,
M.-Y.
,
Gordon
,
A.
,
Thete
,
M. V.
,
Valencia
,
A. M.
,
Revah
,
O.
,
Paşca
,
A. M.
,
Geschwind
,
D. H.
and
Paşca
,
S. P.
(
2022
).
Dissecting the molecular basis of human interneuron migration in forebrain assembloids from Timothy syndrome
.
Cell Stem Cell
29
,
248
-
264.e7
.
Bittman
,
K.
,
Owens
,
D. F.
,
Kriegstein
,
A. R.
and
Lo Turco
,
J. J.
(
1997
).
Cell coupling and uncoupling in the ventricular zone of developing neocortex
.
J. Neurosci.
17
,
7037
-
7044
.
Blackiston
,
D. J.
,
McLaughlin
,
K. A.
and
Levin
,
M.
(
2009
).
Bioelectric controls of cell proliferation: ion channels, membrane voltage and the cell cycle
.
Cell Cycle
8
,
3527
-
3536
.
Blanquie
,
O.
,
Liebmann
,
L.
,
Hübner
,
C. A.
,
Luhmann
,
H. J.
and
Sinning
,
A.
(
2017a
).
NKCC1-mediated GABAergic signaling promotes postnatal cell death in neocortical Cajal-Retzius cells
.
Cereb. Cortex
27
,
1644
-
1659
.
Blanquie
,
O.
,
Yang
,
J.-W.
,
Kilb
,
W.
,
Sharopov
,
S.
,
Sinning
,
A.
and
Luhmann
,
H. J.
(
2017b
).
Electrical activity controls area-specific expression of neuronal apoptosis in the mouse developing cerebral cortex
.
eLife
6
,
e27696
.
Bonnefont
,
J.
and
Vanderhaeghen
,
P.
(
2021
).
Neuronal fate acquisition and specification: time for a change
.
Curr. Opin. Neurobiol.
66
,
195
-
204
.
Bonni
,
A.
,
Sun
,
Y.
,
Nadal-Vicens
,
M.
,
Bhatt
,
A.
,
Frank
,
D. A.
,
Rozovsky
,
I.
,
Stahl
,
N.
,
Yancopoulos
,
G. D.
and
Greenberg
,
M. E.
(
1997
).
Regulation of gliogenesis in the central nervous system by the JAK-STAT signaling pathway
.
Science
278
,
477
-
483
.
Borodinsky
,
L. N.
and
Spitzer
,
N. C.
(
2007
).
Activity-dependent neurotransmitter-receptor matching at the neuromuscular junction
.
Proc. Natl. Acad. Sci. USA
104
,
335
-
340
.
Bortone
,
D.
and
Polleux
,
F.
(
2009
).
KCC2 expression promotes the termination of cortical interneuron migration in a voltage-sensitive calcium-dependent manner
.
Neuron
62
,
53
-
71
.
Boulting
,
G. L.
,
Durresi
,
E.
,
Ataman
,
B.
,
Sherman
,
M. A.
,
Mei
,
K.
,
Harmin
,
D. A.
,
Carter
,
A. C.
,
Hochbaum
,
D. R.
,
Granger
,
A. J.
,
Engreitz
,
J. M.
et al. 
(
2021
).
Activity-dependent regulome of human GABAergic neurons reveals new patterns of gene regulation and neurological disease heritability
.
Nat. Neurosci.
24
,
437
-
448
.
Brandt
,
P.
and
Neve
,
R. L.
(
1992
).
Expression of plasma membrane calcium-pumping ATPase mRNAs in developing rat brain and adult brain subregions: evidence for stage-specific expression
.
J. Neurochem.
59
,
1566
-
1569
.
Brown
,
E. M.
,
Gamba
,
G.
,
Riccardi
,
D.
,
Lombardi
,
M.
,
Butters
,
R.
,
Kifor
,
O.
,
Sun
,
A.
,
Hediger
,
M. A.
,
Lytton
,
J.
and
Hebert
,
S. C.
(
1993
).
Cloning and characterization of an extracellular Ca(2+)-sensing receptor from bovine parathyroid
.
Nature
366
,
575
-
580
.
Buchsbaum
,
I. Y.
and
Cappello
,
S.
(
2019
).
Neuronal migration in the CNS during development and disease: insights from in vivo and in vitro models
.
Development
146
,
dev163766
.
Callaway
,
E. M.
and
Borrell
,
V.
(
2011
).
Developmental sculpting of dendritic morphology of layer 4 neurons in visual cortex: influence of retinal input
.
J. Neurosci.
31
,
7456
-
7470
.
Cancedda
,
L.
,
Fiumelli
,
H.
,
Chen
,
K.
and
Poo
,
M.-M.
(
2007
).
Excitatory GABA action is essential for morphological maturation of cortical neurons in vivo
.
J. Neurosci.
27
,
5224
-
5235
.
Carafoli
,
E.
,
Genazzani
,
A.
and
Guerini
,
D.
(
1999
).
Calcium controls the transcription of its own transporters and channels in developing neurons
.
Biochem. Biophys. Res. Commun.
266
,
624
-
632
.
Carvalho
,
L. D. S.
,
Teixeira
,
L. K.
,
Carrossini
,
N.
,
Caldeira
,
A. T. N.
,
Ansel
,
K. M.
,
Rao
,
A.
and
Viola
,
J. P. B.
(
2007
).
The NFAT1 transcription factor is a repressor of cyclin A2 gene expression
.
Cell Cycle
6
,
1789
-
1795
.
Catterall
,
W. A.
(
2011
).
Voltage-gated calcium channels
.
Cold Spring Harb. Perspect. Biol.
3
,
a003947
.
Cebolla
,
B.
,
Fernández-Pérez
,
A.
,
Perea
,
G.
,
Araque
,
A.
and
Vallejo
,
M.
(
2008
).
DREAM mediates cAMP-dependent, Ca2+-induced stimulation of GFAP gene expression and regulates cortical astrogliogenesis
.
J. Neurosci.
28
,
6703
-
6713
.
Cheli
,
V. T.
,
Santiago González
,
D. A.
,
Spreuer
,
V.
and
Paez
,
P. M.
(
2015
).
Voltage-gated Ca++ entry promotes oligodendrocyte progenitor cell maturation and myelination in vitro
.
Exp. Neurol
265
,
69
-
83
.
Cheli
,
V. T.
,
Santiago González
,
D. A.
,
Namgyal Lama
,
T.
,
Spreuer
,
V.
,
Handley
,
V.
,
Murphy
,
G. G.
and
Paez
,
P. M.
(
2016
).
Conditional deletion of the L-type calcium channel Cav1.2 in oligodendrocyte progenitor cells affects postnatal myelination in mice
.
J. Neurosci.
36
,
10853
-
10869
.
Chen
,
Y.
and
Ghosh
,
A.
(
2005
).
Regulation of dendritic development by neuronal activity
.
J. Neurobiol.
64
,
4
-
10
.
Chen
,
M.
,
Wang
,
J.
,
Jiang
,
J.
,
Zheng
,
X.
,
Justice
,
N. J.
,
Wang
,
K.
,
Ran
,
X.
,
Li
,
Y.
,
Huo
,
Q.
,
Zhang
,
J.
et al. 
(
2017
).
APP modulates KCC2 expression and function in hippocampal GABAergic inhibition
.
eLife
6
,
e20142
.
Ciceri
,
G.
,
Cho
,
H.
,
Kshirsagar
,
M.
,
Baggiolini
,
A.
,
Aromolaran
,
K. A.
,
Walsh
,
R. M.
,
Goldstein
,
P. A.
,
Koche
,
R. P.
,
Leslie
,
C. S.
and
Studer
,
L.
(
2022
).
An epigenetic barrier sets the timing of human neuronal maturation
.
bioRxiv
,
2022.06.02.490114
.
Clapham
,
D. E.
(
2007
).
Calcium signaling
.
Cell
131
,
1047
-
1058
.
Clark
,
M. B.
,
Wrzesinski
,
T.
,
Garcia
,
A. B.
,
Hall
,
N. A. L.
,
Kleinman
,
J. E.
,
Hyde
,
T.
,
Weinberger
,
D. R.
,
Harrison
,
P. J.
,
Haerty
,
W.
and
Tunbridge
,
E. M.
(
2020
).
Long-read sequencing reveals the complex splicing profile of the psychiatric risk gene CACNA1C in human brain
.
Mol. Psychiatry
25
,
37
-
47
.
Close
,
J.
,
Xu
,
H.
,
De Marco García
,
N.
,
Batista-Brito
,
R.
,
Rossignol
,
E.
,
Rudy
,
B.
and
Fishell
,
G.
(
2012
).
Satb1 is an activity-modulated transcription factor required for the terminal differentiation and connectivity of medial ganglionic eminence-derived cortical interneurons
.
J. Neurosci.
32
,
17690
-
17705
.
Contractor
,
A.
,
Ethell
,
I. M.
and
Portera-Cailliau
,
C.
(
2021
).
Cortical interneurons in autism
.
Nat. Neurosci.
24
,
1648
-
1659
.
Cook
,
E. H.
, Jr
,
Courchesne
,
R. Y.
,
Cox
,
N. J.
,
Lord
,
C.
,
Gonen
,
D.
,
Guter
,
S. J.
,
Lincoln
,
A.
,
Nix
,
K.
,
Haas
,
R.
,
Leventhal
,
B. L.
et al. 
(
1998
).
Linkage-disequilibrium mapping of autistic disorder, with 15q11-13 markers
.
Am. J. Hum. Genet
62
,
1077
-
1083
.
Côté
,
F.
,
Fligny
,
C.
,
Bayard
,
E.
,
Launay
,
J.-M.
,
Gershon
,
M. D.
,
Mallet
,
J.
and
Vodjdani
,
G.
(
2007
).
Maternal serotonin is crucial for murine embryonic development
.
Proc. Natl. Acad. Sci. U. S. A
104
,
329
-
334
.
Couturier
,
C. P.
,
Ayyadhury
,
S.
,
Le
,
P. U.
,
Nadaf
,
J.
,
Monlong
,
J.
,
Riva
,
G.
,
Allache
,
R.
,
Baig
,
S.
,
Yan
,
X.
,
Bourgey
,
M.
et al. 
(
2020
).
Single-cell RNA-seq reveals that glioblastoma recapitulates a normal neurodevelopmental hierarchy
.
Nat. Commun.
11
,
3406
.
Cross-Disorder Group of the Psychiatric Genomics Consortium
(
2013
).
Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis
.
Lancet
381
,
1371
-
1379
.
Cunningham
,
C. L.
,
Martínez-Cerdeño
,
V.
and
Noctor
,
S. C.
(
2013
).
Microglia regulate the number of neural precursor cells in the developing cerebral cortex
.
J. Neurosci.
33
,
4216
-
4233
.
Cuzon
,
V. C.
,
Yeh
,
P. W.
,
Cheng
,
Q.
and
Yeh
,
H. H.
(
2006
).
Ambient GABA promotes cortical entry of tangentially migrating cells derived from the medial ganglionic eminence
.
Cereb. Cortex
16
,
1377
-
1388
.
Darbellay
,
B.
,
Arnaudeau
,
S.
,
Bader
,
C. R.
,
Konig
,
S.
and
Bernheim
,
L.
(
2011
).
STIM1L is a new actin-binding splice variant involved in fast repetitive Ca2+ release
.
J. Cell Biol.
194
,
335
-
346
.
De Biase
,
L. M.
,
Nishiyama
,
A.
and
Bergles
,
D. E.
(
2010
).
Excitability and synaptic communication within the oligodendrocyte lineage
.
J. Neurosci.
30
,
3600
-
3611
.
Dehay
,
C.
and
Kennedy
,
H.
(
2007
).
Cell-cycle control and cortical development
.
Nat. Rev. Neurosci.
8
,
438
-
450
.
Deidda
,
G.
,
Parrini
,
M.
,
Naskar
,
S.
,
Bozarth
,
I. F.
,
Contestabile
,
A.
and
Cancedda
,
L.
(
2015
).
Reversing excitatory GABAAR signaling restores synaptic plasticity and memory in a mouse model of Down syndrome
.
Nat. Med.
21
,
318
-
326
.
Deisseroth
,
K.
,
Bito
,
H.
and
Tsien
,
R. W.
(
1996
).
Signaling from synapse to nucleus: postsynaptic CREB phosphorylation during multiple forms of hippocampal synaptic plasticity
.
Neuron
16
,
89
-
101
.
De Marco García
,
N. V.
,
Karayannis
,
T.
and
Fishell
,
G.
(
2011
).
Neuronal activity is required for the development of specific cortical interneuron subtypes
.
Nature
472
,
351
-
355
.
Demerens
,
C.
,
Stankoff
,
B.
,
Logak
,
M.
,
Anglade
,
P.
,
Allinquant
,
B.
,
Couraud
,
F.
,
Zalc
,
B.
and
Lubetzki
,
C.
(
1996
).
Induction of myelination in the central nervous system by electrical activity
.
Proc. Natl. Acad. Sci. U. S. A
93
,
9887
-
9892
.
Denaxa
,
M.
,
Kalaitzidou
,
M.
,
Garefalaki
,
A.
,
Achimastou
,
A.
,
Lasrado
,
R.
,
Maes
,
T.
and
Pachnis
,
V.
(
2012
).
Maturation-promoting activity of SATB1 in MGE-derived cortical interneurons
.
Cell Rep.
2
,
1351
-
1362
.
Denaxa
,
M.
,
Neves
,
G.
,
Rabinowitz
,
A.
,
Kemlo
,
S.
,
Liodis
,
P.
,
Burrone
,
J.
and
Pachnis
,
V.
(
2018
).
Modulation of apoptosis controls inhibitory interneuron number in the cortex
.
Cell Rep.
22
,
1710
-
1721
.
De Rubeis
,
S.
,
He
,
X.
,
Goldberg
,
A. P.
,
Poultney
,
C. S.
,
Samocha
,
K.
,
Cicek
,
A. E.
,
Kou
,
Y.
,
Liu
,
L.
,
Fromer
,
M.
,
Walker
,
S.
et al. 
(
2014
).
Synaptic, transcriptional and chromatin genes disrupted in autism
.
Nature
515
,
209
-
215
.
Dolmetsch
,
R. E.
,
Lewis
,
R. S.
,
Goodnow
,
C. C.
and
Healy
,
J. I.
(
1997
).
Differential activation of transcription factors induced by Ca2+ response amplitude and duration
.
Nature
386
,
855
-
858
.
Dolmetsch
,
R. E.
,
Xu
,
K.
and
Lewis
,
R. S.
(
1998
).
Calcium oscillations increase the efficiency and specificity of gene expression
.
Nature
392
,
933
-
936
.
Dolmetsch
,
R. E.
,
Pajvani
,
U.
,
Fife
,
K.
,
Spotts
,
J. M.
and
Greenberg
,
M. E.
(
2001
).
Signaling to the nucleus by an L-type calcium channel-calmodulin complex through the MAP kinase pathway
.
Science
294
,
333
-
339
.
Dooley
,
A. E.
,
Pappas
,
I. S.
and
Parnavelas
,
J. G.
(
1997
).
Serotonin promotes the survival of cortical glutamatergic neurons in vitro
.
Exp. Neurol.
148
,
205
-
214
.
Doshina
,
A.
,
Gourgue
,
F.
,
Onizuka
,
M.
,
Opsomer
,
R.
,
Wang
,
P.
,
Ando
,
K.
,
Tasiaux
,
B.
,
Dewachter
,
I.
,
Kienlen-Campard
,
P.
,
Brion
,
J.-P.
et al. 
(
2017
).
Cortical cells reveal APP as a new player in the regulation of GABAergic neurotransmission
.
Sci. Rep.
7
,
370
.
Duan
,
Z. R. S.
,
Che
,
A.
,
Chu
,
P.
,
Modol
,
L.
,
Bollmann
,
Y.
,
Babij
,
R.
,
Fetcho
,
R. N.
,
Otsuka
,
T.
,
Fuccillo
,
M. V.
,
Liston
,
C.
et al. 
(
2020
).
GABAergic restriction of network dynamics regulates interneuron survival in the developing cortex
.
Neuron
105
,
75
-
92.e5
.
Echevarria-Cooper
,
D. M.
,
Hawkins
,
N. A.
,
Misra
,
S. N.
,
Huffman
,
A. M.
,
Thaxton
,
T.
,
Thompson
,
C. H.
,
Ben-Shalom
,
R.
,
Nelson
,
A. D.
,
Lipkin
,
A. M.
,
George
,
A. L.
et al. 
(
2022
).
Cellular and behavioral effects of altered NaV1.2 sodium channel ion permeability in Scn2aK1422E mice
.
Hum. Mol. Genet.
[Epub ahead of print].
Endele
,
S.
,
Rosenberger
,
G.
,
Geider
,
K.
,
Popp
,
B.
,
Tamer
,
C.
,
Stefanova
,
I.
,
Milh
,
M.
,
Kortüm
,
F.
,
Fritsch
,
A.
,
Pientka
,
F. K.
et al. 
(
2010
).
Mutations in GRIN2A and GRIN2B encoding regulatory subunits of NMDA receptors cause variable neurodevelopmental phenotypes
.
Nat. Genet.
42
,
1021
-
1026
.
Erreger
,
K.
,
Dravid
,
S. M.
,
Banke
,
T. G.
,
Wyllie
,
D. J. A.
and
Traynelis
,
S. F.
(
2005
).
Subunit-specific gating controls rat NR1/NR2A and NR1/NR2B NMDA channel kinetics and synaptic signalling profiles
.
J. Physiol.
563
,
345
-
358
.
Fame
,
R. M.
and
Lehtinen
,
M. K.
(
2020
).
Emergence and developmental roles of the cerebrospinal fluid system
.
Dev. Cell
52
,
261
-
275
.
Favuzzi
,
E.
,
Huang
,
S.
,
Saldi
,
G. A.
,
Binan
,
L.
,
Ibrahim
,
L. A.
,
Fernández-Otero
,
M.
,
Cao
,
Y.
,
Zeine
,
A.
,
Sefah
,
A.
,
Zheng
,
K.
et al. 
(
2021
).
GABA-receptive microglia selectively sculpt developing inhibitory circuits
.
Cell
184
,
4048
-
4063
.e32.
Ferrer
,
I.
,
Barrachina
,
M.
,
Puig
,
B.
,
Martínez de Lagrán
,
M.
,
Martí
,
E.
,
Avila
,
J.
and
Dierssen
,
M.
(
2005
).
Constitutive Dyrk1A is abnormally expressed in Alzheimer disease, Down syndrome, Pick disease, and related transgenic models
.
Neurobiol. Dis.
20
,
392
-
400
.
Fietz
,
S. A.
,
Kelava
,
I.
,
Vogt
,
J.
,
Wilsch-Bräuninger
,
M.
,
Stenzel
,
D.
,
Fish
,
J. L.
,
Corbeil
,
D.
,
Riehn
,
A.
,
Distler
,
W.
,
Nitsch
,
R.
et al. 
(
2010
).
OSVZ progenitors of human and ferret neocortex are epithelial-like and expand by integrin signaling
.
Nat. Neurosci.
13
,
690
-
699
.
Fiumelli
,
H.
,
Cancedda
,
L.
and
Poo
,
M.-M.
(
2005
).
Modulation of GABAergic transmission by activity via postsynaptic Ca2+-dependent regulation of KCC2 function
.
Neuron
48
,
773
-
786
.
Flint
,
A. C.
,
Liu
,
X.
and
Kriegstein
,
A. R.
(
1998
).
Nonsynaptic glycine receptor activation during early neocortical development
.
Neuron
20
,
43
-
53
.
Foster
,
T. C.
,
Sharrow
,
K. M.
,
Masse
,
J. R.
,
Norris
,
C. M.
and
Kumar
,
A.
(
2001
).
Calcineurin links Ca2+ dysregulation with brain aging
.
J. Neurosci
21
,
4066
-
4073
.
Francis
,
F.
and
Cappello
,
S.
(
2021
).
Neuronal migration and disorders – an update
.
Curr. Opin. Neurobiol.
66
,
57
-
68
.
Fukushima
,
M.
,
Tomita
,
T.
,
Janoshazi
,
A.
and
Putney
,
J. W.
(
2012
).
Alternative translation initiation gives rise to two isoforms of Orai1 with distinct plasma membrane mobilities
.
J. Cell Sci
125
,
4354
-
4361
.
Fulton
,
D.
,
Paez
,
P. M.
,
Fisher
,
R.
,
Handley
,
V.
,
Colwell
,
C. S.
and
Campagnoni
,
A. T.
(
2010
).
Regulation of L-type Ca++ currents and process morphology in white matter oligodendrocyte precursor cells by golli-myelin proteins
.
Glia
58
,
1292
-
1303
.
Furman
,
J. L.
,
Sama
,
D. M.
,
Gant
,
J. C.
,
Beckett
,
T. L.
,
Paul Murphy
,
M.
,
Bachstetter
,
A. D.
,
Van Eldik
,
L. J.
and
Norris
,
C. M.
(
2012
).
Targeting Astrocytes Ameliorates Neurologic Changes in a Mouse Model of Alzheimer's Disease
.
J. Neurosci
32
,
16129
-
16140
.
Furukawa
,
T.
,
Yamada
,
J.
,
Akita
,
T.
,
Matsushima
,
Y.
,
Yanagawa
,
Y.
and
Fukuda
,
A.
(
2014
).
Roles of taurine-mediated tonic GABAA receptor activation in the radial migration of neurons in the fetal mouse cerebral cortex
.
Front. Cell. Neurosci
8
,
88
.
Gandal
,
M. J.
,
Zhang
,
P.
,
Hadjimichael
,
E.
,
Walker
,
R. L.
,
Chen
,
C.
,
Liu
,
S.
,
Won
,
H.
,
van Bakel
,
H.
,
Varghese
,
M.
,
Wang
,
Y.
et al. 
(
2018
).
Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder
.
Science
362
,
eaat8127
.
Ganguly
,
K.
,
Schinder
,
A. F.
,
Wong
,
S. T.
and
Poo
,
M.
(
2001
).
GABA itself promotes the developmental switch of neuronal GABAergic responses from excitation to inhibition
.
Cell
105
,
521
-
532
.
Gary
,
D. S.
,
Malone
,
M.
,
Capestany
,
P.
,
Houdayer
,
T.
and
McDonald
,
J. W.
(
2012
).
Electrical stimulation promotes the survival of oligodendrocytes in mixed cortical cultures
.
J. Neurosci. Res.
90
,
72
-
83
.
Gaudillière
,
B.
,
Konishi
,
Y.
,
de la Iglesia
,
N.
,
Yao
,
G. L.
and
Bonni
,
A.
(
2004
).
A CaMKII-NeuroD signaling pathway specifies dendritic morphogenesis
.
Neuron
41
,
229
-
241
.
Ghosh
,
A.
,
Carnahan
,
J.
and
Greenberg
,
M. E.
(
1994
).
Requirement for BDNF in activity-dependent survival of cortical neurons
.
Science
263
,
1618
-
1623
.
Gibson
,
E. M.
,
Purger
,
D.
,
Mount
,
C. W.
,
Goldstein
,
A. K.
,
Lin
,
G. L.
,
Wood
,
L. S.
,
Inema
,
I.
,
Miller
,
S. E.
,
Bieri
,
G.
,
Zuchero
,
J. B.
et al. 
(
2014
).
Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain
.
Science
344
,
1252304
.
Ginhoux
,
F.
,
Greter
,
M.
,
Leboeuf
,
M.
,
Nandi
,
S.
,
See
,
P.
,
Gokhan
,
S.
,
Mehler
,
M. F.
,
Conway
,
S. J.
,
Ng
,
L. G.
,
Stanley
,
E. R.
et al. 
(
2010
).
Fate mapping analysis reveals that adult microglia derive from primitive macrophages
.
Science
330
,
841
-
845
.
Golbs
,
A.
,
Nimmervoll
,
B.
,
Sun
,
J.-J.
,
Sava
,
I. E.
and
Luhmann
,
H. J.
(
2011
).
Control of programmed cell death by distinct electrical activity patterns
.
Cereb. Cortex
21
,
1192
-
1202
.
Gomez
,
T. M.
and
Spitzer
,
N. C.
(
1999
).
In vivo regulation of axon extension and pathfinding by growth-cone calcium transients
.
Nature
397
,
350
-
355
.
Gomez
,
T. M.
and
Zheng
,
J. Q.
(
2006
).
The molecular basis for calcium-dependent axon pathfinding
.
Nat. Rev. Neurosci.
7
,
115
-
125
.
Graef
,
I. A.
,
Mermelstein
,
P. G.
,
Stankunas
,
K.
,
Neilson
,
J. R.
,
Deisseroth
,
K.
,
Tsien
,
R. W.
and
Crabtree
,
G. R.
(
1999
).
L-type calcium channels and GSK-3 regulate the activity of NF-ATc4 in hippocampal neurons
.
Nature
401
,
703
-
708
.
Greer
,
P. L.
and
Greenberg
,
M. E.
(
2008
).
From synapse to nucleus: calcium-dependent gene transcription in the control of synapse development and function
.
Neuron
59
,
846
-
860
.
Gu
,
X.
and
Spitzer
,
N. C.
(
1995
).
Distinct aspects of neuronal differentiation encoded by frequency of spontaneous Ca2+ transients
.
Nature
375
,
784
-
787
.
Gu
,
X.
,
Olson
,
E. C.
and
Spitzer
,
N. C.
(
1994
).
Spontaneous neuronal calcium spikes and waves during early differentiation
.
J. Neurosci
14
,
6325
-
6335
.
Hansen
,
D. V.
,
Lui
,
J. H.
,
Parker
,
P. R. L.
and
Kriegstein
,
A. R.
(
2010
).
Neurogenic radial glia in the outer subventricular zone of human neocortex
.
Nature
464
,
554
-
561
.
Haubensak
,
W.
,
Attardo
,
A.
,
Denk
,
W.
and
Huttner
,
W. B.
(
2004
).
Neurons arise in the basal neuroepithelium of the early mammalian telencephalon: a major site of neurogenesis
.
Proc. Natl. Acad. Sci. U. S. A
101
,
3196
-
3201
.
Haydar
,
T. F.
,
Wang
,
F.
,
Schwartz
,
M. L.
and
Rakic
,
P.
(
2000
).
Differential modulation of proliferation in the neocortical ventricular and subventricular zones
.
J. Neurosci.
20
,
5764
-
5774
.
He
,
S.
,
Ma
,
J.
,
Liu
,
N.
and
Yu
,
X.
(
2010
).
Early enriched environment promotes neonatal GABAergic neurotransmission and accelerates synapse maturation
.
J. Neurosci
30
,
7910
-
7916
.
Heck
,
N.
,
Kilb
,
W.
,
Reiprich
,
P.
,
Kubota
,
H.
,
Furukawa
,
T.
,
Fukuda
,
A.
and
Luhmann
,
H. J.
(
2007
).
GABA-A receptors regulate neocortical neuronal migration in vitro and in vivo
.
Cereb. Cortex
17
,
138
-
148
.
Heck
,
N.
,
Golbs
,
A.
,
Riedemann
,
T.
,
Sun
,
J.-J.
,
Lessmann
,
V.
and
Luhmann
,
H. J.
(
2008
).
Activity-dependent regulation of neuronal apoptosis in neonatal mouse cerebral cortex
.
Cereb. Cortex
18
,
1335
-
1349
.
Heck
,
A.
,
Fastenrath
,
M.
,
Coynel
,
D.
,
Auschra
,
B.
,
Bickel
,
H.
,
Freytag
,
V.
,
Gschwind
,
L.
,
Hartmann
,
F.
,
Jessen
,
F.
,
Kaduszkiewicz
,
H.
et al. 
(
2015
).
Genetic analysis of association between calcium signaling and hippocampal activation, memory performance in the young and old, and risk for sporadic Alzheimer disease
.
JAMA Psychiatry
72
,
1029
-
1036
.
Henson
,
M. A.
,
Roberts
,
A. C.
,
Salimi
,
K.
,
Vadlamudi
,
S.
,
Hamer
,
R. M.
,
Gilmore
,
J. H.
,
Jarskog
,
L. F.
and
Philpot
,
B. D.
(
2008
).
Developmental regulation of the NMDA receptor subunits, NR3A and NR1, in human prefrontal cortex
.
Cereb. Cortex
18
,
2560
-
2573
.
Hergenreder
,
E.
,
Zorina
,
Y.
,
Zhao
,
Z.
,
Munguba
,
H.
,
Calder
,
E. L.
,
Baggiolini
,
A.
,
Minotti
,
A. P.
,
Walsh
,
R. M.
,
Liston
,
C.
,
Levitz
,
J.
et al. 
(
2022
).
Combined small molecule treatment accelerates timing of maturation in human pluripotent stem cell-derived neurons
.
bioRxiv
,
2022.06.02.494616
.
Hirabayashi
,
Y.
,
Kwon
,
S.-K.
,
Paek
,
H.
,
Pernice
,
W. M.
,
Paul
,
M. A.
,
Lee
,
J.
,
Erfani
,
P.
,
Raczkowski
,
A.
,
Petrey
,
D. S.
,
Pon
,
L. A.
et al. 
(
2017
).
ER-mitochondria tethering by PDZD8 regulates Ca2+ dynamics in mammalian neurons
.
Science
358
,
623
-
630
.
Hirai
,
K.
,
Yoshioka
,
H.
,
Kihara
,
M.
,
Hasegawa
,
K.
,
Sakamoto
,
T.
,
Sawada
,
T.
and
Fushiki
,
S.
(
1999
).
Inhibiting neuronal migration by blocking NMDA receptors in the embryonic rat cerebral cortex: a tissue culture study
.
Brain Res. Dev. Brain Res.
114
,
63
-
67
.
Hoerder-Suabedissen
,
A.
and
Molnár
,
Z.
(
2015
).
Development, evolution and pathology of neocortical subplate neurons
.
Nat. Rev. Neurosci.
16
,
133
-
146
.
Holt
,
C. E.
,
Martin
,
K. C.
and
Schuman
,
E. M.
(
2019
).
Local translation in neurons: visualization and function
.
Nat. Struct. Mol. Biol.
26
,
557
-
566
.
Huang
,
T.
,
Xie
,
Z.
,
Wang
,
J.
,
Li
,
M.
,
Jing
,
N.
and
Li
,
L.
(
2011
).
Nuclear factor of activated T Cells (NFAT) proteins repress canonical Wnt signaling via its interaction with dishevelled (Dvl) protein and participate in regulating neural progenitor cell proliferation and differentiation *
.
J. Biol. Chem.
286
,
37399
-
37405
.
Hughes
,
A. N.
and
Appel
,
B.
(
2020
).
Microglia phagocytose myelin sheaths to modify developmental myelination
.
Nat. Neurosci.
23
,
1055
-
1066
.
Ikonomidou
,
C.
,
Bosch
,
F.
,
Miksa
,
M.
,
Bittigau
,
P.
,
Vöckler
,
J.
,
Dikranian
,
K.
,
Tenkova
,
T. I.
,
Stefovska
,
V.
,
Turski
,
L.
and
Olney
,
J. W.
(
1999
).
Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain
.
Science
283
,
70
-
74
.
Imamura
,
K.
,
Sahara
,
N.
,
Kanaan
,
N. M.
,
Tsukita
,
K.
,
Kondo
,
T.
,
Kutoku
,
Y.
,
Ohsawa
,
Y.
,
Sunada
,
Y.
,
Kawakami
,
K.
,
Hotta
,
A.
et al. 
(
2016
).
Calcium dysregulation contributes to neurodegeneration in FTLD patient iPSC-derived neurons
.
Sci. Rep.
6
,
1
-
10
.
Iossifov
,
I.
,
O'Roak
,
B. J.
,
Sanders
,
S. J.
,
Ronemus
,
M.
,
Krumm
,
N.
,
Levy
,
D.
,
Stessman
,
H. A.
,
Witherspoon
,
K. T.
,
Vives
,
L.
,
Patterson
,
K. E.
et al. 
(
2014
).
The contribution of de novo coding mutations to autism spectrum disorder
.
Nature
515
,
216
-
221
.
Ivashko-Pachima
,
Y.
,
Hadar
,
A.
,
Grigg
,
I.
,
Korenková
,
V.
,
Kapitansky
,
O.
,
Karmon
,
G.
,
Gershovits
,
M.
,
Sayas
,
C. L.
,
Kooy
,
R. F.
,
Attems
,
J.
et al. 
(
2021
).
Discovery of autism/intellectual disability somatic mutations in Alzheimer's brains: mutated ADNP cytoskeletal impairments and repair as a case study
.
Mol. Psychiatry
26
,
1619
-
1633
.
Iwasato
,
T.
,
Datwani
,
A.
,
Wolf
,
A. M.
,
Nishiyama
,
H.
,
Taguchi
,
Y.
,
Tonegawa
,
S.
,
Knöpfel
,
T.
,
Erzurumlu
,
R. S.
and
Itohara
,
S.
(
2000
).
Cortex-restricted disruption of NMDAR1 impairs neuronal patterns in the barrel cortex
.
Nature
406
,
726
-
731
.
Ji
,
F.
,
Kanbara
,
N.
and
Obata
,
K.
(
1999
).
GABA and histogenesis in fetal and neonatal mouse brain lacking both the isoforms of glutamic acid decarboxylase
.
Neurosci. Res.
33
,
187
-
194
.
Jiang
,
H.
,
Jiang
,
W.
,
Zou
,
J.
,
Wang
,
B.
,
Yu
,
M.
,
Pan
,
Y.
,
Lin
,
Y.
,
Mao
,
Y.
and
Wang
,
Y.
(
2015
).
The GluN2B subunit of N-methy-D-asparate receptor regulates the radial migration of cortical neurons in vivo
.
Brain Res.
1610
,
20
-
32
.
Kamijo
,
S.
,
Ishii
,
Y.
,
Horigane
,
S.-I.
,
Suzuki
,
K.
,
Ohkura
,
M.
,
Nakai
,
J.
,
Fujii
,
H.
,
Takemoto-Kimura
,
S.
and
Bito
,
H.
(
2018
).
A critical neurodevelopmental role for L-Type voltage-gated calcium channels in neurite extension and radial migration
.
J. Neurosci.
38
,
5551
-
5566
.
Kandler
,
K.
and
Katz
,
L. C.
(
1998
).
Coordination of neuronal activity in developing visual cortex by gap junction-mediated biochemical communication
.
J. Neurosci.
18
,
1419
-
1427
.
Kanold
,
P. O.
(
2019
).
The first cortical circuits: Subplate neurons lead the way and shape cortical organization
.
Neuroforum
25
,
15
-
23
.
Kanold
,
P. O.
and
Shatz
,
C. J.
(
2006
).
Subplate neurons regulate maturation of cortical inhibition and outcome of ocular dominance plasticity
.
Neuron
51
,
627
-
638
.
Kessaris
,
N.
,
Fogarty
,
M.
,
Iannarelli
,
P.
,
Grist
,
M.
,
Wegner
,
M.
and
Richardson
,
W. D.
(
2006
).
Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage
.
Nat. Neurosci.
9
,
173
-
179
.
Khan
,
T. A.
,
Revah
,
O.
,
Gordon
,
A.
,
Yoon
,
S.-J.
,
Krawisz
,
A. K.
,
Goold
,
C.
,
Sun
,
Y.
,
Kim
,
C. H.
,
Tian
,
Y.
,
Li
,
M.-Y.
et al. 
(
2020
).
Neuronal defects in a human cellular model of 22q11.2 deletion syndrome
.
Nat. Med.
26
,
1888
-
1898
.
Kip
,
S. N.
,
Gray
,
N. W.
,
Burette
,
A.
,
Canbay
,
A.
,
Weinberg
,
R. J.
and
Strehler
,
E. E.
(
2006
).
Changes in the expression of plasma membrane calcium extrusion systems during the maturation of hippocampal neurons
.
Hippocampus
16
,
20
-
34
.
Kline
,
C. F.
,
Scott
,
J.
,
Curran
,
J.
,
Hund
,
T. J.
and
Mohler
,
P. J.
(
2014
).
Ankyrin-B regulates Cav2.1 and Cav2.2 channel expression and targeting
.
J. Biol. Chem
289
,
5285
-
5295
.
Komuro
,
H.
and
Rakic
,
P.
(
1992
).
Selective role of N-type calcium channels in neuronal migration
.
Science
257
,
806
-
809
.
Komuro
,
H.
and
Rakic
,
P.
(
1993
).
Modulation of neuronal migration by NMDA receptors
.
Science
260
,
95
-
97
.
Krasnow
,
A. M.
,
Ford
,
M. C.
,
Valdivia
,
L. E.
,
Wilson
,
S. W.
and
Attwell
,
D.
(
2018
).
Regulation of developing myelin sheath elongation by oligodendrocyte calcium transients in vivo
.
Nat. Neurosci.
21
,
24
-
28
.
Krey
,
J. F.
,
Paşca
,
S. P.
,
Shcheglovitov
,
A.
,
Yazawa
,
M.
,
Schwemberger
,
R.
,
Rasmusson
,
R.
and
Dolmetsch
,
R. E.
(
2013
).
Timothy syndrome is associated with activity-dependent dendritic retraction in rodent and human neurons
.
Nat. Neurosci.
16
,
201
-
209
.
Kumar
,
S. S.
,
Bacci
,
A.
,
Kharazia
,
V.
and
Huguenard
,
J. R.
(
2002
).
A developmental switch of AMPA receptor subunits in neocortical pyramidal neurons
.
J. Neurosci.
22
,
3005
-
3015
.
Lewis
,
R. S.
(
2011
).
Store-operated calcium channels: new perspectives on mechanism and function
.
Cold Spring Harb. Perspect. Biol
3
,
a003970
.
Lewis
,
T. L.
,
Kwon
,
S.-K.
,
Lee
,
A.
,
Shaw
,
R.
and
Polleux
,
F.
(
2018
).
MFF-dependent mitochondrial fission regulates presynaptic release and axon branching by limiting axonal mitochondria size
.
Nat. Commun.
9
,
1
-
15
.
Li
,
H.
,
Fertuzinhos
,
S.
,
Mohns
,
E.
,
Hnasko
,
T. S.
,
Verhage
,
M.
,
Edwards
,
R.
,
Sestan
,
N.
and
Crair
,
M. C.
(
2013
).
Laminar and columnar development of barrel cortex relies on thalamocortical neurotransmission
.
Neuron
79
,
970
-
986
.
Libé-Philippot
,
B.
and
Vanderhaeghen
,
P.
(
2021
).
Cellular and molecular mechanisms linking human cortical development and evolution
.
Annu. Rev. Genet.
55
,
555
-
581
.
Lim
,
L.
,
Mi
,
D.
,
Llorca
,
A.
and
Marín
,
O.
(
2018
).
Development and functional diversification of cortical interneurons
.
Neuron
100
,
294
-
313
.
Lin
,
J. H.-C.
,
Takano
,
T.
,
Arcuino
,
G.
,
Wang
,
X.
,
Hu
,
F.
,
Darzynkiewicz
,
Z.
,
Nunes
,
M.
,
Goldman
,
S. A.
and
Nedergaard
,
M.
(
2007
).
Purinergic signaling regulates neural progenitor cell expansion and neurogenesis
.
Dev. Biol.
302
,
356
-
366
.
Lin
,
Y.
,
Bloodgood
,
B. L.
,
Hauser
,
J. L.
,
Lapan
,
A. D.
,
Koon
,
A. C.
,
Kim
,
T.-K.
,
Hu
,
L. S.
,
Malik
,
A. N.
and
Greenberg
,
M. E.
(
2008
).
Activity-dependent regulation of inhibitory synapse development by Npas4
.
Nature
455
,
1198
-
1204
.
Lin
,
Y.
,
Yang
,
J.
,
Shen
,
Z.
,
Ma
,
J.
,
Simons
,
B. D.
and
Shi
,
S.-H.
(
2021
).
Behavior and lineage progression of neural progenitors in the mammalian cortex
.
Curr. Opin. Neurobiol.
66
,
144
-
157
.
Lipscombe
,
D.
,
Andrade
,
A.
and
Allen
,
S. E.
(
2013
).
Alternative splicing: functional diversity among voltage-gated calcium channels and behavioral consequences
.
Biochim. Biophys. Acta
1828
,
1522
-
1529
.
Liu
,
L.
,
Cavanaugh
,
J. E.
,
Wang
,
Y.
,
Sakagami
,
H.
,
Mao
,
Z.
and
Xia
,
Z.
(
2003
).
ERK5 activation of MEF2-mediated gene expression plays a critical role in BDNF-promoted survival of developing but not mature cortical neurons
.
Proc. Natl. Acad. Sci. U. S. A
100
,
8532
-
8537
.
Liu
,
X.
,
Hashimoto-Torii
,
K.
,
Torii
,
M.
,
Haydar
,
T. F.
and
Rakic
,
P.
(
2008
).
The role of ATP signaling in the migration of intermediate neuronal progenitors to the neocortical subventricular zone
.
Proc. Natl. Acad. Sci. U. S. A
105
,
11802
-
11807
.
Liu
,
X.
,
Hashimoto-Torii
,
K.
,
Torii
,
M.
,
Ding
,
C.
and
Rakic
,
P.
(
2010
).
Gap junctions/hemichannels modulate interkinetic nuclear migration in the forebrain precursors
.
J. Neurosci
30
,
4197
-
4209
.
Llorca
,
A.
and
Marín
,
O.
(
2021
).
Orchestrated freedom: new insights into cortical neurogenesis
.
Curr. Opin. Neurobiol
66
,
48
-
56
.
López-Bendito
,
G.
(
2018
).
Development of the thalamocortical interactions: past, present and future
.
Neuroscience
385
,
67
-
74
.
López-Bendito
,
G.
,
Shigemoto
,
R.
,
Kulik
,
A.
,
Paulsen
,
O.
,
Fairén
,
A.
and
Luján
,
R.
(
2002
).
Expression and distribution of metabotropic GABA receptor subtypes GABABR1 and GABABR2 during rat neocortical development
.
Eur. J. Neurosci.
15
,
1766
-
1778
.
Lott
,
I. T.
and
Head
,
E.
(
2019
).
Dementia in Down syndrome: unique insights for Alzheimer disease research
.
Nat. Rev. Neurol
15
,
135
-
147
.
Lo Turco
,
L.
and
Kriegstein
,
J. J.
(
1991
).
Clusters of coupled neuroblasts in embryonic neocortex
.
Science
252
,
563
-
566
.
Lo Turco
,
J. J.
,
Blanton
,
M. G.
and
Kriegstein
,
A. R.
(
1991
).
Initial expression and endogenous activation of NMDA channels in early neocortical development
.
J. Neurosci.
11
,
792
-
799
.
Lo Turco
,
J. J.
,
Owens
,
D. F.
,
Heath
,
M. J.
,
Davis
,
M. B.
and
Kriegstein
,
A. R.
(
1995
).
GABA and glutamate depolarize cortical progenitor cells and inhibit DNA synthesis
.
Neuron
15
,
1287
-
1298
.
Ludwig
,
A.
,
Uvarov
,
P.
,
Soni
,
S.
,
Thomas-Crusells
,
J.
,
Airaksinen
,
M. S.
and
Rivera
,
C.
(
2011
).
Early growth response 4 mediates BDNF induction of potassium chloride cotransporter 2 transcription
.
J. Neurosci.
31
,
644
-
649
.
Luhmann
,
H. J.
,
Fukuda
,
A.
and
Kilb
,
W.
(
2015
).
Control of cortical neuronal migration by glutamate and GABA
.
Front. Cell. Neurosci
9
,
4
.
Luo
,
F.
,
Sclip
,
A.
,
Jiang
,
M.
and
Südhof
,
T. C.
(
2020
).
Neurexins cluster Ca2+ channels within the presynaptic active zone
.
EMBO J.
39
,
e103208
.
Lyons
,
M. R.
and
West
,
A. E.
(
2011
).
Mechanisms of specificity in neuronal activity-regulated gene transcription
.
Prog. Neurobiol
94
,
259
-
295
.
Ma
,
W.
,
Maric
,
D.
,
Li
,
B. S.
,
Hu
,
Q.
,
Andreadis
,
J. D.
,
Grant
,
G. M.
,
Liu
,
Q. Y.
,
Shaffer
,
K. M.
,
Chang
,
Y. H.
,
Zhang
,
L.
et al. 
(
2000
).
Acetylcholine stimulates cortical precursor cell proliferation in vitro via muscarinic receptor activation and MAP kinase phosphorylation
.
Eur. J. Neurosci.
12
,
1227
-
1240
.
Ma
,
H.
,
Groth
,
R. D.
,
Cohen
,
S. M.
,
Emery
,
J. F.
,
Li
,
B.
,
Hoedt
,
E.
,
Zhang
,
G.
,
Neubert
,
T. A.
and
Tsien
,
R. W.
(
2014
).
γCaMKII shuttles Ca2+/CaM to the nucleus to trigger CREB phosphorylation and gene expression
.
Cell
159
,
281
-
294
.
Malmersjö
,
S.
,
Rebellato
,
P.
,
Smedler
,
E.
,
Planert
,
H.
,
Kanatani
,
S.
,
Liste
,
I.
,
Nanou
,
E.
,
Sunner
,
H.
,
Abdelhady
,
S.
,
Zhang
,
S.
et al. 
(
2013
).
Neural progenitors organize in small-world networks to promote cell proliferation
.
Proc. Natl. Acad. Sci. U. S. A
110
,
E1524
-
E1532
.
Mao
,
Z.
,
Bonni
,
A.
,
Xia
,
F.
,
Nadal-Vicens
,
M.
and
Greenberg
,
M. E.
(
1999
).
Neuronal activity-dependent cell survival mediated by transcription factor MEF2
.
Science
286
,
785
-
790
.
Maric
,
D.
,
Liu
,
Q. Y.
,
Grant
,
G. M.
,
Andreadis
,
J. D.
,
Hu
,
Q.
,
Chang
,
Y. H.
,
Barker
,
J. L.
,
Joseph
,
J.
,
Stenger
,
D. A.
and
Ma
,
W.
(
2000
).
Functional ionotropic glutamate receptors emerge during terminal cell division and early neuronal differentiation of rat neuroepithelial cells
.
J. Neurosci. Res.
61
,
652
-
662
.
Marisca
,
R.
,
Hoche
,
T.
,
Agirre
,
E.
,
Hoodless
,
L. J.
,
Barkey
,
W.
,
Auer
,
F.
,
Castelo-Branco
,
G.
and
Czopka
,
T.
(
2020
).
Functionally distinct subgroups of oligodendrocyte precursor cells integrate neural activity and execute myelin formation
.
Nat. Neurosci.
23
,
363
-
374
.
Martini
,
F. J.
and
Valdeolmillos
,
M.
(
2010
).
Actomyosin contraction at the cell rear drives nuclear translocation in migrating cortical interneurons
.
J. Neurosci.
30
,
8660
-
8670
.
Mayer
,
C.
,
Hafemeister
,
C.
,
Bandler
,
R. C.
,
Machold
,
R.
,
Batista Brito
,
R.
,
Jaglin
,
X.
,
Allaway
,
K.
,
Butler
,
A.
,
Fishell
,
G.
and
Satija
,
R.
(
2018
).
Developmental diversification of cortical inhibitory interneurons
.
Nature
555
,
457
-
462
.
Mayer
,
S.
,
Chen
,
J.
,
Velmeshev
,
D.
,
Mayer
,
A.
,
Eze
,
U. C.
,
Bhaduri
,
A.
,
Cunha
,
C. E.
,
Jung
,
D.
,
Arjun
,
A.
,
Li
,
E.
et al. 
(
2019
).
Multimodal single-cell analysis reveals physiological maturation in the developing human neocortex
.
Neuron
102
,
143
-
158.e7
.
McAllister
,
A. K.
,
Katz
,
L. C.
and
Lo
,
D. C.
(
1996
).
Neurotrophin regulation of cortical dendritic growth requires activity
.
Neuron
17
,
1057
-
1064
.
McManus
,
M. F.
,
Chen
,
L. C.
,
Vallejo
,
I.
and
Vallejo
,
M.
(
1999
).
Astroglial differentiation of cortical precursor cells triggered by activation of the cAMP-dependent signaling pathway
.
J. Neurosci
19
,
9004
-
9015
.
Medina
,
D. L.
,
Di Paola
,
S.
,
Peluso
,
I.
,
Armani
,
A.
,
De Stefani
,
D.
,
Venditti
,
R.
,
Montefusco
,
S.
,
Scotto-Rosato
,
A.
,
Prezioso
,
C.
,
Forrester
,
A.
et al. 
(
2015
).
Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB
.
Nat. Cell Biol.
17
,
288
-
299
.
Medvedeva
,
V. P.
and
Pierani
,
A.
(
2020
).
How do electric fields coordinate neuronal migration and maturation in the developing cortex?
Front. Cell Dev. Biol.
8
,
580657
.
Mencer
,
S.
,
Kartawy
,
M.
,
Lendenfeld
,
F.
,
Soluh
,
H.
,
Tripathi
,
M. K.
,
Khaliulin
,
I.
and
Amal
,
H.
(
2021
).
Proteomics of autism and Alzheimer's mouse models reveal common alterations in mTOR signaling pathway
.
Transl. Psychiatry
11
,
480
.
Messersmith
,
E. K.
,
Feller
,
M. B.
,
Zhang
,
H.
and
Shatz
,
C. J.
(
1997
).
Migration of neocortical neurons in the absence of functional NMDA receptors
.
Mol. Cell. Neurosci.
9
,
347
-
357
.
Métin
,
C.
,
Denizot
,
J. P.
and
Ropert
,
N.
(
2000
).
Intermediate zone cells express calcium-permeable AMPA receptors and establish close contact with growing axons
.
J. Neurosci
20
,
696
-
708
.
Miller
,
F. D.
and
Gauthier
,
A. S.
(
2007
).
Timing is everything: making neurons versus glia in the developing cortex
.
Neuron
54
,
357
-
369
.
Missler
,
M.
,
Zhang
,
W.
,
Rohlmann
,
A.
,
Kattenstroth
,
G.
,
Hammer
,
R. E.
,
Gottmann
,
K.
and
Südhof
,
T. C.
(
2003
).
Alpha-neurexins couple Ca2+ channels to synaptic vesicle exocytosis
.
Nature
423
,
939
-
948
.
Mitew
,
S.
,
Gobius
,
I.
,
Fenlon
,
L. R.
,
McDougall
,
S. J.
,
Hawkes
,
D.
,
Xing
,
Y. L.
,
Bujalka
,
H.
,
Gundlach
,
A. L.
,
Richards
,
L. J.
,
Kilpatrick
,
T. J.
et al. 
(
2018
).
Pharmacogenetic stimulation of neuronal activity increases myelination in an axon-specific manner
.
Nat. Commun.
9
,
306
.
Mizuno
,
H.
,
Hirano
,
T.
and
Tagawa
,
Y.
(
2007
).
Evidence for activity-dependent cortical wiring: formation of interhemispheric connections in neonatal mouse visual cortex requires projection neuron activity
.
J. Neurosci
27
,
6760
-
6770
.
Molnár
,
Z.
,
Luhmann
,
H. J.
and
Kanold
,
P. O.
(
2020
).
Transient cortical circuits match spontaneous and sensory-driven activity during development
.
Science
370
,
eabb2153
.
Monyer
,
H.
,
Burnashev
,
N.
,
Laurie
,
D. J.
,
Sakmann
,
B.
and
Seeburg
,
P. H.
(
1994
).
Developmental and regional expression in the rat brain and functional properties of four NMDA receptors
.
Neuron
12
,
529
-
540
.
Murase
,
S.
,
Owens
,
D. F.
and
McKay
,
R. D.
(
2011
).
In the newborn hippocampus, neurotrophin-dependent survival requires spontaneous activity and integrin signaling
.
J. Neurosci.
31
,
7791
-
7800
.
Murthy
,
S.
,
Niquille
,
M.
,
Hurni
,
N.
,
Limoni
,
G.
,
Frazer
,
S.
,
Chameau
,
P.
,
van Hooft
,
J. A.
,
Vitalis
,
T.
and
Dayer
,
A.
(
2014
).
Serotonin receptor 3A controls interneuron migration into the neocortex
.
Nat. Commun.
5
,
1
-
10
.
Nimmervoll
,
B.
,
Denter
,
D. G.
,
Sava
,
I.
,
Kilb
,
W.
and
Luhmann
,
H. J.
(
2011
).
Glycine receptors influence radial migration in the embryonic mouse neocortex
.
Neuroreport
22
,
509
-
513
.
Nishimoto
,
M.
,
Furuta
,
A.
,
Aoki
,
S.
,
Kudo
,
Y.
,
Miyakawa
,
H.
and
Wada
,
K.
(
2007
).
PACAP/PAC1 autocrine system promotes proliferation and astrogenesis in neural progenitor cells
.
Glia
55
,
317
-
327
.
Noctor
,
S. C.
,
Martínez-Cerdeño
,
V.
,
Ivic
,
L.
and
Kriegstein
,
A. R.
(
2004
).
Cortical neurons arise in symmetric and asymmetric division zones and migrate through specific phases
.
Nat. Neurosci.
7
,
136
-
144
.
Norris
,
C. M.
,
Kadish
,
I.
,
Blalock
,
E. M.
,
Chen
,
K.-C.
,
Thibault
,
V.
,
Porter
,
N. M.
,
Landfield
,
P. W.
and
Kraner
,
S. D.
(
2005
).
Calcineurin triggers reactive/inflammatory processes in astrocytes and is upregulated in aging and Alzheimer's models
.
J. Neurosci.
25
,
4649
-
4658
.
O'Hare
,
J. K.
,
Gonzalez
,
K. C.
,
Herrlinger
,
S. A.
,
Hirabayashi
,
Y.
,
Hewitt
,
V. L.
,
Blockus
,
H.
,
Szoboszlay
,
M.
,
Rolotti
,
S. V.
,
Geiller
,
T. C.
,
Negrean
,
A.
et al. 
(
2022
).
Compartment-specific tuning of dendritic feature selectivity by intracellular Ca2+ release
.
Science
375
,
eabm1670
.
Ohtaka-Maruyama
,
C.
,
Okamoto
,
M.
,
Endo
,
K.
,
Oshima
,
M.
,
Kaneko
,
N.
,
Yura
,
K.
,
Okado
,
H.
,
Miyata
,
T.
and
Maeda
,
N.
(
2018
).
Synaptic transmission from subplate neurons controls radial migration of neocortical neurons
.
Science
360
,
313
-
317
.
Oliveria
,
S. F.
,
Dell'Acqua
,
M. L.
and
Sather
,
W. A.
(
2007
).
AKAP79/150 anchoring of calcineurin controls neuronal L-type Ca2+ channel activity and nuclear signaling
.
Neuron
55
,
261
-
275
.
Oliveria
,
S. F.
,
Dittmer
,
P. J.
,
Youn
,
D.-H.
,
Dell'Acqua
,
M. L.
and
Sather
,
W. A.
(
2012
).
Localized calcineurin confers Ca2+-dependent inactivation on neuronal L-type Ca2+ channels
.
J. Neurosci
32
,
15328
-
15337
.
Osswald
,
M.
,
Jung
,
E.
,
Sahm
,
F.
,
Solecki
,
G.
,
Venkataramani
,
V.
,
Blaes
,
J.
,
Weil
,
S.
,
Horstmann
,
H.
,
Wiestler
,
B.
,
Syed
,
M.
et al. 
(
2015
).
Brain tumour cells interconnect to a functional and resistant network
.
Nature
528
,
93
-
98
.
Owens
,
D. F.
and
Kriegstein
,
A. R.
(
1998
).
Patterns of intracellular calcium fluctuation in precursor cells of the neocortical ventricular zone
.
J. Neurosci.
18
,
5374
-
5388
.
Owens
,
D. F.
,
Boyce
,
L. H.
,
Davis
,
M. B.
and
Kriegstein
,
A. R.
(
1996
).
Excitatory GABA responses in embryonic and neonatal cortical slices demonstrated by gramicidin perforated-patch recordings and calcium imaging
.
J. Neurosci.
16
,
6414
-
6423
.
Owens
,
D. F.
,
Liu
,
X.
and
Kriegstein
,
A. R.
(
1999
).
Changing properties of GABA(A) receptor-mediated signaling during early neocortical development
.
J. Neurophysiol
82
,
570
-
583
.
Owens
,
D. F.
,
Flint
,
A. C.
,
Dammerman
,
R. S.
and
Kriegstein
,
A. R.
(
2000
).
Calcium dynamics of neocortical ventricular zone cells
.
Dev. Neurosci.
22
,
25
-
33
.
Pan
,
Y.
and
Monje
,
M.
(
2020
).
Activity shapes neural circuit form and function: a historical perspective
.
J. Neurosci
40
,
944
-
954
.
Panagiotakos
,
G.
and
Pasca
,
S. P.
(
2022
).
A matter of space and time: emerging roles of disease-associated proteins in neural development
.
Neuron
110
,
195
-
208
.
Panagiotakos
,
G.
,
Haveles
,
C.
,
Arjun
,
A.
,
Petrova
,
R.
,
Rana
,
A.
,
Portmann
,
T.
,
Paşca
,
S. P.
,
Palmer
,
T. D.
and
Dolmetsch
,
R. E.
(
2019
).
Aberrant calcium channel splicing drives defects in cortical differentiation in Timothy syndrome
.
eLife
8
,
e51037
.
Paoletti
,
P.
,
Bellone
,
C.
and
Zhou
,
Q.
(
2013
).
NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease
.
Nat. Rev. Neurosci.
14
,
383
-
400
.
Parekh
,
A. B.
(
2008
).
Ca2+ microdomains near plasma membrane Ca2+ channels: impact on cell function
.
J. Physiol.
586
,
3043
-
3054
.
Parikshak
,
N. N.
,
Swarup
,
V.
,
Belgard
,
T. G.
,
Irimia
,
M.
,
Ramaswami
,
G.
,
Gandal
,
M. J.
,
Hartl
,
C.
,
Leppa
,
V.
,
Ubieta
,
L. d. l. T.
,
Huang
,
J.
et al. 
(
2016
).
Genome-wide changes in lncRNA, splicing, and regional gene expression patterns in autism
.
Nature
540
,
423
-
427
.
Park
,
C. Y.
,
Shcheglovitov
,
A.
and
Dolmetsch
,
R.
(
2010
).
The CRAC channel activator STIM1 binds and inhibits L-type voltage-gated calcium channels
.
Science
330
,
101
-
105
.
Park
,
J.
,
Wetzel
,
I.
,
Marriott
,
I.
,
Dréau
,
D.
,
D'Avanzo
,
C.
,
Kim
,
D. Y.
,
Tanzi
,
R. E.
and
Cho
,
H.
(
2018
).
A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer's disease
.
Nat. Neurosci
21
,
941
-
951
.
Paşca
,
S. P.
,
Portmann
,
T.
,
Voineagu
,
I.
,
Yazawa
,
M.
,
Shcheglovitov
,
A.
,
Paşca
,
A. M.
,
Cord
,
B.
,
Palmer
,
T. D.
,
Chikahisa
,
S.
,
Nishino
,
S.
et al. 
(
2011
).
Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome
.
Nat. Med.
17
,
1657
-
1662
.
Pchitskaya
,
E.
,
Popugaeva
,
E.
and
Bezprozvanny
,
I.
(
2018
).
Calcium signaling and molecular mechanisms underlying neurodegenerative diseases
.
Cell Calcium
70
,
87
-
94
.
Polleux
,
F.
,
Whitford
,
K. L.
,
Dijkhuizen
,
P. A.
,
Vitalis
,
T.
and
Ghosh
,
A.
(
2002
).
Control of cortical interneuron migration by neurotrophins and PI3-kinase signaling
.
Development
129
,
3147
-
3160
.
Prakriya
,
M.
and
Lewis
,
R. S.
(
2015
).
Store-operated calcium channels
.
Physiol. Rev.
95
,
1383
-
1436
.
Priya
,
R.
,
Paredes
,
M. F.
,
Karayannis
,
T.
,
Yusuf
,
N.
,
Liu
,
X.
,
Jaglin
,
X.
,
Graef
,
I.
,
Alvarez-Buylla
,
A.
and
Fishell
,
G.
(
2018
).
Activity regulates cell death within cortical interneurons through a Calcineurin-dependent mechanism
.
Cell Rep.
22
,
1695
-
1709
.
Puehringer
,
D.
,
Orel
,
N.
,
Lüningschrör
,
P.
,
Subramanian
,
N.
,
Herrmann
,
T.
,
Chao
,
M. V.
and
Sendtner
,
M.
(
2013
).
EGF transactivation of Trk receptors regulates the migration of newborn cortical neurons
.
Nat. Neurosci.
16
,
407
-
415
.
Putney
,
J. W.
,
Steinckwich-Besançon
,
N.
,
Numaga-Tomita
,
T.
,
Davis
,
F. M.
,
Desai
,
P. N.
,
D'Agostin
,
D. M.
,
Wu
,
S.
and
Bird
,
G. S.
(
2017
).
The functions of store-operated calcium channels
.
Biochim. Biophys. Acta Mol. Cell Res.
1864
,
900
-
906
.
Qian
,
X.
,
Davis
,
A. A.
,
Goderie
,
S. K.
and
Temple
,
S.
(
1997
).
FGF2 concentration regulates the generation of neurons and glia from multipotent cortical stem cells
.
Neuron
18
,
81
-
93
.
Quadrato
,
G.
,
Benevento
,
M.
,
Alber
,
S.
,
Jacob
,
C.
,
Floriddia
,
E. M.
,
Nguyen
,
T.
,
Elnaggar
,
M. Y.
,
Pedroarena
,
C. M.
,
Molkentin
,
J. D.
and
Di Giovanni
,
S.
(
2012
).
Nuclear factor of activated T cells (NFATc4) is required for BDNF-dependent survival of adult-born neurons and spatial memory formation in the hippocampus
.
Proc. Natl. Acad. Sci. U. S. A
109
,
E1499
-
E1508
.
Quesnel-Vallières
,
M.
,
Dargaei
,
Z.
,
Irimia
,
M.
,
Gonatopoulos-Pournatzis
,
T.
,
Ip
,
J. Y.
,
Wu
,
M.
,
Sterne-Weiler
,
T.
,
Nakagawa
,
S.
,
Woodin
,
M. A.
,
Blencowe
,
B. J.
et al. 
(
2016
).
Misregulation of an activity-dependent splicing network as a common mechanism underlying autism spectrum disorders
.
Mol. Cell
64
,
1023
-
1034
.
Ramesh
,
G.
,
Jarzembowski
,
L.
,
Schwarz
,
Y.
,
Poth
,
V.
,
Konrad
,
M.
,
Knapp
,
M. L.
,
Schwär
,
G.
,
Lauer
,
A. A.
,
Grimm
,
M. O. W.
,
Alansary
,
D.
et al. 
(
2021
).
A short isoform of STIM1 confers frequency-dependent synaptic enhancement
.
Cell Rep.
34
,
108844
.
Rash
,
B. G.
,
Ackman
,
J. B.
and
Rakic
,
P.
(
2016
).
Bidirectional radial Ca(2+) activity regulates neurogenesis and migration during early cortical column formation
.
Sci. Adv.
2
,
e1501733
.
Rash
,
B. G.
,
Micali
,
N.
,
Huttner
,
A. J.
,
Morozov
,
Y. M.
,
Horvath
,
T. L.
and
Rakic
,
P.
(
2018
).
Metabolic regulation and glucose sensitivity of cortical radial glial cells
.
Proc. Natl. Acad. Sci. U. S. A
115
,
10142
-
10147
.
Redmond
,
L.
,
Kashani
,
A. H.
and
Ghosh
,
A.
(
2002
).
Calcium regulation of dendritic growth via CaM kinase IV and CREB-mediated transcription
.
Neuron
34
,
999
-
1010
.
Reiprich
,
P.
,
Kilb
,
W.
and
Luhmann
,
H. J.
(
2005
).
Neonatal NMDA receptor blockade disturbs neuronal migration in rat somatosensory cortex in vivo
.
Cereb. Cortex
15
,
349
-
358
.
Riva
,
M.
,
Genescu
,
I.
,
Habermacher
,
C.
,
Orduz
,
D.
,
Ledonne
,
F.
,
Rijli
,
F. M.
,
López-Bendito
,
G.
,
Coppola
,
E.
,
Garel
,
S.
,
Angulo
,
M. C.
et al. 
(
2019
).
Activity-dependent death of transient Cajal-Retzius neurons is required for functional cortical wiring
.
eLife
8
,
e50503
.
Rivera
,
C.
,
Voipio
,
J.
,
Thomas-Crusells
,
J.
,
Li
,
H.
,
Emri
,
Z.
,
Sipilä
,
S.
,
Payne
,
J. A.
,
Minichiello
,
L.
,
Saarma
,
M.
and
Kaila
,
K.
(
2004
).
Mechanism of activity-dependent downregulation of the neuron-specific K-Cl cotransporter KCC2
.
J. Neurosci
24
,
4683
-
4691
.
Rodríguez-Tornos
,
F. M.
,
Briz
,
C. G.
,
Weiss
,
L. A.
,
Sebastián-Serrano
,
A.
,
Ares
,
S.
,
Navarrete
,
M.
,
Frangeul
,
L.
,
Galazo
,
M.
,
Jabaudon
,
D.
,
Esteban
,
J. A.
et al. 
(
2016
).
Cux1 enables interhemispheric connections of layer II/III neurons by regulating Kv1-dependent firing
.
Neuron
89
,
494
-
506
.
Rosenberg
,
S. S.
and
Spitzer
,
N. C.
(
2011
).
Calcium signaling in neuronal development
.
Cold Spring Harb. Perspect. Biol
3
,
a004259
.
Sanchez-Priego
,
C.
,
Hu
,
R.
,
Boshans
,
L. L.
,
Lalli
,
M.
,
Janas
,
J. A.
,
Williams
,
S. E.
,
Dong
,
Z.
and
Yang
,
N.
(
2022
).
Mapping cis-regulatory elements in human neurons links psychiatric disease heritability and activity-regulated transcriptional programs
.
Cell Rep
39
,
110877
.
Sanders
,
S. J.
,
Campbell
,
A. J.
,
Cottrell
,
J. R.
,
Moller
,
R. S.
,
Wagner
,
F. F.
,
Auldridge
,
A. L.
,
Bernier
,
R. A.
,
Catterall
,
W. A.
,
Chung
,
W. K.
,
Empfield
,
J. R.
et al. 
(
2018
).
Progress in understanding and treating SCN2A-mediated disorders
.
Trends Neurosci.
41
,
442
-
456
.
Schafer
,
D. P.
,
Lehrman
,
E. K.
,
Kautzman
,
A. G.
,
Koyama
,
R.
,
Mardinly
,
A. R.
,
Yamasaki
,
R.
,
Ransohoff
,
R. M.
,
Greenberg
,
M. E.
,
Barres
,
B. A.
and
Stevens
,
B.
(
2012
).
Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner
.
Neuron
74
,
691
-
705
.
Schmunk
,
G.
,
Boubion
,
B. J.
,
Smith
,
I. F.
,
Parker
,
I.
and
Gargus
,
J. J.
(
2015
).
Shared functional defect in IP3R-mediated calcium signaling in diverse monogenic autism syndromes
.
Transl. Psychiatry
5
,
e643
.
Schmunk
,
G.
,
Nguyen
,
R. L.
,
Ferguson
,
D. L.
,
Kumar
,
K.
,
Parker
,
I.
and
Gargus
,
J. J.
(
2017
).
High-throughput screen detects calcium signaling dysfunction in typical sporadic autism spectrum disorder
.
Sci. Rep.
7
,
40740
.
Sernagor
,
E.
,
Young
,
C.
and
Eglen
,
S. J.
(
2003
).
Developmental modulation of retinal wave dynamics: shedding light on the GABA saga
.
J. Neurosci
23
,
7621
-
7629
.
Serrano-Pérez
,
M. C.
,
Fernández
,
M.
,
Neria
,
F.
,
Berjón-Otero
,
M.
,
Doncel-Pérez
,
E.
,
Cano
,
E.
and
Tranque
,
P.
(
2015
).
NFAT transcription factors regulate survival, proliferation, migration, and differentiation of neural precursor cells
.
Glia
63
,
987
-
1004
.
Sheng
,
M.
,
Cummings
,
J.
,
Roldan
,
L. A.
,
Jan
,
Y. N.
and
Jan
,
L. Y.
(
1994
).
Changing subunit composition of heteromeric NMDA receptors during development of rat cortex
.
Nature
368
,
144
-
147
.
Silva
,
C. G.
,
Peyre
,
E.
and
Nguyen
,
L.
(
2019
).
Cell migration promotes dynamic cellular interactions to control cerebral cortex morphogenesis
.
Nat. Rev. Neurosci.
20
,
318
-
329
.
Sohal
,
V. S.
and
Rubenstein
,
J. L. R.
(
2019
).
Excitation-inhibition balance as a framework for investigating mechanisms in neuropsychiatric disorders
.
Mol. Psychiatry
24
,
1248
-
1257
.
Soldatov
,
N. M.
(
1994
).
Genomic structure of human L-type Ca2+ channel
.
Genomics
22
,
77
-
87
.
Soldatov
,
N. M.
,
Zühlke
,
R. D.
,
Bouron
,
A.
and
Reuter
,
H.
(
1997
).
Molecular structures involved in L-type calcium channel inactivation. Role of the carboxyl-terminal region encoded by exons 40–42 in alpha1C subunit in the kinetics and Ca2+ dependence of inactivation
.
J. Biol. Chem.
272
,
3560
-
3566
.
Somasundaram
,
A.
,
Shum
,
A. K.
,
McBride
,
H. J.
,
Kessler
,
J. A.
,
Feske
,
S.
,
Miller
,
R. J.
and
Prakriya
,
M.
(
2014
).
Store-operated CRAC channels regulate gene expression and proliferation in neural progenitor cells
.
J. Neurosci
34
,
9107
-
9123
.
Sommer
,
B.
,
Köhler
,
M.
,
Sprengel
,
R.
and
Seeburg
,
P. H.
(
1991
).
RNA editing in brain controls a determinant of ion flow in glutamate-gated channels
.
Cell
67
,
11
-
19
.
Song
,
M.-R.
and
Ghosh
,
A.
(
2004
).
FGF2-induced chromatin remodeling regulates CNTF-mediated gene expression and astrocyte differentiation
.
Nat. Neurosci.
7
,
229
-
235
.
Soria
,
J. M.
and
Valdeolmillos
,
M.
(
2002
).
Receptor-activated calcium signals in tangentially migrating cortical cells
.
Cereb. Cortex
12
,
831
-
839
.
Southwell
,
D. G.
,
Paredes
,
M. F.
,
Galvao
,
R. P.
,
Jones
,
D. L.
,
Froemke
,
R. C.
,
Sebe
,
J. Y.
,
Alfaro-Cervello
,
C.
,
Tang
,
Y.
,
Garcia-Verdugo
,
J. M.
,
Rubenstein
,
J. L.
et al. 
(
2012
).
Intrinsically determined cell death of developing cortical interneurons
.
Nature
491
,
109
-
113
.
Spitzer
,
S. O.
,
Sitnikov
,
S.
,
Kamen
,
Y.
,
Evans
,
K. A.
,
Kronenberg-Versteeg
,
D.
,
Dietmann
,
S.
,
de Faria
,
O.
, Jr
,
Agathou
,
S.
and
Káradóttir
,
R. T.
(
2019
).
Oligodendrocyte progenitor cells become regionally diverse and heterogeneous with age
.
Neuron
101
,
459
-
471.e5
.
Splawski
,
I.
,
Timothy
,
K. W.
,
Sharpe
,
L. M.
,
Decher
,
N.
,
Kumar
,
P.
,
Bloise
,
R.
,
Napolitano
,
C.
,
Schwartz
,
P. J.
,
Joseph
,
R. M.
,
Condouris
,
K.
et al. 
(
2004
).
Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism
.
Cell
119
,
19
-
31
.
Squarzoni
,
P.
,
Oller
,
G.
,
Hoeffel
,
G.
,
Pont-Lezica
,
L.
,
Rostaing
,
P.
,
Low
,
D.
,
Bessis
,
A.
,
Ginhoux
,
F.
and
Garel
,
S.
(
2014
).
Microglia modulate wiring of the embryonic forebrain
.
Cell Rep.
8
,
1271
-
1279
.
Suárez
,
R.
,
Fenlon
,
L. R.
,
Marek
,
R.
,
Avitan
,
L.
,
Sah
,
P.
,
Goodhill
,
G. J.
and
Richards
,
L. J.
(
2014
).
Balanced interhemispheric cortical activity is required for correct targeting of the corpus callosum
.
Neuron
82
,
1289
-
1298
.
Sun
,
C.
,
Nold
,
A.
,
Fusco
,
C. M.
,
Rangaraju
,
V.
,
Tchumatchenko
,
T.
,
Heilemann
,
M.
and
Schuman
,
E. M.
(
2021
).
The prevalence and specificity of local protein synthesis during neuronal synaptic plasticity
.
Sci. Adv.
7
,
eabj0790
.
Takechi
,
H.
,
Eilers
,
J.
and
Konnerth
,
A.
(
1998
).
A new class of synaptic response involving calcium release in dendritic spines
.
Nature
396
,
757
-
760
.
Tanaka
,
D. H.
,
Maekawa
,
K.
,
Yanagawa
,
Y.
,
Obata
,
K.
and
Murakami
,
F.
(
2006
).
Multidirectional and multizonal tangential migration of GABAergic interneurons in the developing cerebral cortex
.
Development
133
,
2167
-
2176
.
Tang
,
Z. Z.
,
Liang
,
M. C.
,
Lu
,
S.
,
Yu
,
D.
,
Yu
,
C. Y.
,
Yue
,
D. T.
and
Soong
,
T. W.
(
2004
).
Transcript scanning reveals novel and extensive splice variations in human l-type voltage-gated calcium channel, Cav1.2 alpha1 subunit
.
J. Biol. Chem.
279
,
44335
-
44343
.
Tang
,
Z. Z.
,
Hong
,
X.
,
Wang
,
J.
and
Soong
,
T. W.
(
2007
).
Signature combinatorial splicing profiles of rat cardiac- and smooth-muscle Cav1.2 channels with distinct biophysical properties
.
Cell Calcium
41
,
417
-
428
.
Tang
,
Z. Z.
,
Zheng
,
S.
,
Nikolic
,
J.
and
Black
,
D. L.
(
2009
).
Developmental control of CaV1.2 L-type calcium channel splicing by Fox proteins
.
Mol. Cell. Biol.
29
,
4757
-
4765
.
Tang
,
Z. Z.
,
Sharma
,
S.
,
Zheng
,
S.
,
Chawla
,
G.
,
Nikolic
,
J.
and
Black
,
D. L.
(
2011
).
Regulation of the mutually exclusive exons 8a and 8 in the CaV1.2 calcium channel transcript by polypyrimidine tract-binding protein
.
J. Biol. Chem
286
,
10007
-
10016
.
Taubes
,
A.
,
Nova
,
P.
,
Zalocusky
,
K. A.
,
Kosti
,
I.
,
Bicak
,
M.
,
Zilberter
,
M. Y.
,
Hao
,
Y.
,
Yoon
,
S. Y.
,
Oskotsky
,
T.
,
Pineda
,
S.
et al. 
(
2021
).
Experimental and real-world evidence supporting the computational repurposing of bumetanide for APOE4-related Alzheimer's disease
.
Nature Aging
1
,
932
-
947
.
Taverna
,
E.
,
Götz
,
M.
and
Huttner
,
W. B.
(
2014
).
The cell biology of neurogenesis: toward an understanding of the development and evolution of the neocortex
.
Annu. Rev. Cell Dev. Biol.
30
,
465
-
502
.
Teixeira
,
L. K.
,
Carrossini
,
N.
,
Sécca
,
C.
,
Kroll
,
J. E.
,
DaCunha
,
D. C.
,
Faget
,
D. V.
,
Carvalho
,
L. D. S.
,
de Souza
,
S. J.
and
Viola
,
J. P. B.
(
2016
).
NFAT1 transcription factor regulates cell cycle progression and cyclin E expression in B lymphocytes
.
Cell Cycle
15
,
2346
-
2359
.
Thion
,
M. S.
and
Garel
,
S.
(
2017
).
On place and time: microglia in embryonic and perinatal brain development
.
Curr. Opin. Neurobiol.
47
,
121
-
130
.
Thion
,
M. S.
and
Garel
,
S.
(
2020
).
Microglial ontogeny, diversity and neurodevelopmental functions
.
Curr. Opin. Genet. Dev
65
,
186
-
194
.
Toth
,
A. B.
,
Shum
,
A. K.
and
Prakriya
,
M.
(
2016
).
Regulation of neurogenesis by calcium signaling
.
Cell Calcium
59
,
124
-
134
.
Traynelis
,
S. F.
,
Wollmuth
,
L. P.
,
McBain
,
C. J.
,
Menniti
,
F. S.
,
Vance
,
K. M.
,
Ogden
,
K. K.
,
Hansen
,
K. B.
,
Yuan
,
H.
,
Myers
,
S. J.
and
Dingledine
,
R.
(
2010
).
Glutamate receptor ion channels: structure, regulation, and function
.
Pharmacol. Rev.
62
,
405
-
496
.
Tropepe
,
V.
,
Sibilia
,
M.
,
Ciruna
,
B. G.
,
Rossant
,
J.
,
Wagner
,
E. F.
and
van der Kooy
,
D.
(
1999
).
Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon
.
Dev. Biol.
208
,
166
-
188
.
Tyssowski
,
K. M.
,
DeStefino
,
N. R.
,
Cho
,
J.-H.
,
Dunn
,
C. J.
,
Poston
,
R. G.
,
Carty
,
C. E.
,
Jones
,
R. D.
,
Chang
,
S. M.
,
Romeo
,
P.
,
Wurzelmann
,
M. K.
et al. 
(
2018
).
Different neuronal activity patterns induce different gene expression programs
.
Neuron
98
,
530
-
546
.
Tyzio
,
R.
,
Cossart
,
R.
,
Khalilov
,
I.
,
Minlebaev
,
M.
,
Hübner
,
C. A.
,
Represa
,
A.
,
Ben-Ari
,
Y.
and
Khazipov
,
R.
(
2006
).
Maternal oxytocin triggers a transient inhibitory switch in GABA signaling in the fetal brain during delivery
.
Science
314
,
1788
-
1792
.
Tyzio
,
R.
,
Nardou
,
R.
,
Ferrari
,
D. C.
,
Tsintsadze
,
T.
,
Shahrokhi
,
A.
,
Eftekhari
,
S.
,
Khalilov
,
I.
,
Tsintsadze
,
V.
,
Brouchoud
,
C.
,
Chazal
,
G.
et al. 
(
2014
).
Oxytocin-mediated GABA inhibition during delivery attenuates autism pathogenesis in rodent offspring
.
Science
343
,
675
-
679
.
Ueno
,
M.
,
Fujita
,
Y.
,
Tanaka
,
T.
,
Nakamura
,
Y.
,
Kikuta
,
J.
,
Ishii
,
M.
and
Yamashita
,
T.
(
2013
).
Layer V cortical neurons require microglial support for survival during postnatal development
.
Nat. Neurosci.
16
,
543
-
551
.
Uhlén
,
P.
,
Fritz
,
N.
,
Smedler
,
E.
,
Malmersjö
,
S.
and
Kanatani
,
S.
(
2015
).
Calcium signaling in neocortical development
.
Dev. Neurobiol
75
,
360
-
368
.
Umpierre
,
A. D.
and
Wu
,
L.-J.
(
2021
).
How microglia sense and regulate neuronal activity
.
Glia
69
,
1637
-
1653
.
Urso
,
K.
,
Fernández
,
A.
,
Velasco
,
P.
,
Cotrina
,
J.
,
de Andrés
,
B.
,
Sánchez-Gómez
,
P.
,
Hernández-Laín
,
A.
,
Hortelano
,
S.
,
Redondo
,
J. M.
and
Cano
,
E.
(
2019
).
NFATc3 controls tumour growth by regulating proliferation and migration of human astroglioma cells
.
Sci. Rep.
9
,
9361
.
Vallano
,
M. L.
,
Beaman-Hall
,
C. M.
and
Benmansour
,
S.
(
1999
).
Ca2+ and pH modulate alternative splicing of exon 5 in NMDA receptor subunit 1
.
Neuroreport
10
,
3659
-
3664
.
Vallejo
,
I.
and
Vallejo
,
M.
(
2002
).
Pituitary adenylate cyclase-activating polypeptide induces astrocyte differentiation of precursor cells from developing cerebral cortex
.
Mol. Cell. Neurosci.
21
,
671
-
683
.
Venkataramani
,
V.
,
Tanev
,
D. I.
,
Strahle
,
C.
,
Studier-Fischer
,
A.
,
Fankhauser
,
L.
,
Kessler
,
T.
,
Körber
,
C.
,
Kardorff
,
M.
,
Ratliff
,
M.
,
Xie
,
R.
et al. 
(
2019
).
Glutamatergic synaptic input to glioma cells drives brain tumour progression
.
Nature
573
,
532
-
538
.
Venkatesh
,
H. S.
,
Morishita
,
W.
,
Geraghty
,
A. C.
,
Silverbush
,
D.
,
Gillespie
,
S. M.
,
Arzt
,
M.
,
Tam
,
L. T.
,
Espenel
,
C.
,
Ponnuswami
,
A.
,
Ni
,
L.
et al. 
(
2019
).
Electrical and synaptic integration of glioma into neural circuits
.
Nature
573
,
539
-
545
.
Venteicher
,
A. S.
,
Tirosh
,
I.
,
Hebert
,
C.
,
Yizhak
,
K.
,
Neftel
,
C.
,
Filbin
,
M. G.
,
Hovestadt
,
V.
,
Escalante
,
L. E.
,
Shaw
,
M. L.
,
Rodman
,
C.
et al. 
(
2017
).
Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq
.
Science
355
,
eaai8478
.
Verkhratsky
,
A.
(
2019
).
Astroglial calcium signaling in aging and Alzheimer's disease
.
Cold Spring Harb. Perspect. Biol.
11
,
035188
.
Vihma
,
H.
,
Pruunsild
,
P.
and
Timmusk
,
T.
(
2008
).
Alternative splicing and expression of human and mouse NFAT genes
.
Genomics
92
,
279
-
291
.
Vitali
,
I.
,
Fièvre
,
S.
,
Telley
,
L.
,
Oberst
,
P.
,
Bariselli
,
S.
,
Frangeul
,
L.
,
Baumann
,
N.
,
McMahon
,
J. J.
,
Klingler
,
E.
,
Bocchi
,
R.
et al. 
(
2018
).
Progenitor hyperpolarization regulates the sequential generation of neuronal subtypes in the developing neocortex
.
Cell
174
,
1264
-
1276.e15
.
Vivanti
,
G.
,
Tao
,
S.
,
Lyall
,
K.
,
Robins
,
D. L.
and
Shea
,
L. L.
(
2021
).
The prevalence and incidence of early-onset dementia among adults with autism spectrum disorder
.
Autism Res.
14
,
2189
-
2199
.
Vo
,
N.
,
Klein
,
M. E.
,
Varlamova
,
O.
,
Keller
,
D. M.
,
Yamamoto
,
T.
,
Goodman
,
R. H.
and
Impey
,
S.
(
2005
).
A cAMP-response element binding protein-induced microRNA regulates neuronal morphogenesis
.
Proc. Natl. Acad. Sci. U. S. A
102
,
16426
-
16431
.
Voigt
,
T.
,
Baier
,
H.
and
Dolabela de Lima
,
A.
(
1997
).
Synchronization of neuronal activity promotes survival of individual rat neocortical neurons in early development
.
Eur. J. Neurosci.
9
,
990
-
999
.
Walker
,
W. H.
,
Girardet
,
C.
and
Habener
,
J. F.
(
1996
).
Alternative exon splicing controls a translational switch from activator to repressor isoforms of transcription factor CREB during spermatogenesis
.
J. Biol. Chem.
271
,
20145
-
21050
.
Wamsley
,
B.
,
Jaglin
,
X. H.
,
Favuzzi
,
E.
,
Quattrocolo
,
G.
,
Nigro
,
M. J.
,
Yusuf
,
N.
,
Khodadadi-Jamayran
,
A.
,
Rudy
,
B.
and
Fishell
,
G.
(
2018
).
Rbfox1 mediates cell-type-specific splicing in cortical interneurons
.
Neuron
100
,
846
-
859.e7
.
Wang
,
C.-L.
,
Zhang
,
L.
,
Zhou
,
Y.
,
Zhou
,
J.
,
Yang
,
X.-J.
,
Duan
,
S.-M.
,
Xiong
,
Z.-Q.
and
Ding
,
Y.-Q.
(
2007
).
Activity-dependent development of callosal projections in the somatosensory cortex
.
J. Neurosci
27
,
11334
-
11342
.
Wang
,
Y.
,
Deng
,
X.
,
Mancarella
,
S.
,
Hendron
,
E.
,
Eguchi
,
S.
,
Soboloff
,
J.
,
Tang
,
X. D.
and
Gill
,
D. L.
(
2010
).
The calcium store sensor, STIM1, reciprocally controls Orai and CaV1.2 channels
.
Science
330
,
105
-
109
.
Wang
,
T.
,
Guo
,
H.
,
Xiong
,
B.
,
Stessman
,
H. A. F.
,
Wu
,
H.
,
Coe
,
B. P.
,
Turner
,
T. N.
,
Liu
,
Y.
,
Zhao
,
W.
,
Hoekzema
,
K.
et al. 
(
2016
).
De novo genic mutations among a Chinese autism spectrum disorder cohort
.
Nat. Commun.
7
,
13316
.
Wang
,
R.
,
Sharma
,
R.
,
Shen
,
X.
,
Laughney
,
A. M.
,
Funato
,
K.
,
Clark
,
P. J.
,
Shpokayte
,
M.
,
Morgenstern
,
P.
,
Navare
,
M.
,
Xu
,
Y.
et al. 
(
2020
).
Adult human glioblastomas harbor radial glia-like cells
.
Stem Cell Rep.
14
,
338
-
350
.
Wayman
,
G. A.
,
Impey
,
S.
,
Marks
,
D.
,
Saneyoshi
,
T.
,
Grant
,
W. F.
,
Derkach
,
V.
and
Soderling
,
T. R.
(
2006
).
Activity-dependent dendritic arborization mediated by CaM-kinase I activation and enhanced CREB-dependent transcription of Wnt-2
.
Neuron
50
,
897
-
909
.
Weissman
,
T. A.
,
Riquelme
,
P. A.
,
Ivic
,
L.
,
Flint
,
A. C.
and
Kriegstein
,
A. R.
(
2004
).
Calcium waves propagate through radial glial cells and modulate proliferation in the developing neocortex
.
Neuron
43
,
647
-
661
.
Welling
,
A.
,
Ludwig
,
A.
,
Zimmer
,
S.
,
Klugbauer
,
N.
,
Flockerzi
,
V.
and
Hofmann
,
F.
(
1997
).
Alternatively spliced IS6 segments of the alpha 1C gene determine the tissue-specific dihydropyridine sensitivity of cardiac and vascular smooth muscle L-type Ca2+ channels
.
Circ. Res
81
,
526
-
532
.
West
,
A. E.
,
Chen
,
W. G.
,
Dalva
,
M. B.
,
Dolmetsch
,
R. E.
,
Kornhauser
,
J. M.
,
Shaywitz
,
A. J.
,
Takasu
,
M. A.
,
Tao
,
X.
and
Greenberg
,
M. E.
(
2001
).
Calcium regulation of neuronal gene expression
.
Proc. Natl. Acad. Sci. U. S. A
98
,
11024
-
11031
.
Whitney
,
N. P.
,
Peng
,
H.
,
Erdmann
,
N. B.
,
Tian
,
C.
,
Monaghan
,
D. T.
and
Zheng
,
J. C.
(
2008
).
Calcium-permeable AMPA receptors containing Q/R-unedited GluR2 direct human neural progenitor cell differentiation to neurons
.
FASEB J.
22
,
2888
-
2900
.
Wild
,
A. R.
,
Sinnen
,
B. L.
,
Dittmer
,
P. J.
,
Kennedy
,
M. J.
,
Sather
,
W. A.
and
Dell'Acqua
,
M. L.
(
2019
).
Synapse-to-nucleus communication through NFAT is mediated by L-type Ca2+ channel Ca2+ spike propagation to the soma
.
Cell Rep.
26
,
3537
-
3550.e4
.
Wong
,
F. K.
,
Bercsenyi
,
K.
,
Sreenivasan
,
V.
,
Portalés
,
A.
,
Fernández-Otero
,
M.
and
Marín
,
O.
(
2018
).
Pyramidal cell regulation of interneuron survival sculpts cortical networks
.
Nature
557
,
668
-
673
.
Wong
,
F. K.
,
Selten
,
M.
,
Rosés-Novella
,
C.
,
Sreenivasan
,
V.
,
Pallas-Bazarra
,
N.
,
Serafeimidou-Pouliou
,
E.
,
Hanusz-Godoy
,
A.
,
Oozeer
,
F.
,
Edwards
,
R.
and
Marín
,
O.
(
2022
).
Serotonergic regulation of bipolar cell survival in the developing cerebral cortex
.
Cell Rep.
40
,
111037
.
Wong Fong Sang
,
I. E.
,
Schroer
,
J.
,
Halbhuber
,
L.
,
Warm
,
D.
,
Yang
,
J.-W.
,
Luhmann
,
H. J.
,
Kilb
,
W.
and
Sinning
,
A.
(
2021
).
Optogenetically controlled activity pattern determines survival rate of developing neocortical neurons
.
Int. J. Mol. Sci.
22
,
6575
.
Wyllie
,
D. J. A.
,
Livesey
,
M. R.
and
Hardingham
,
G. E.
(
2013
).
Influence of GluN2 subunit identity on NMDA receptor function
.
Neuropharmacology
74
,
4
-
17
.
Yang
,
L.
,
Wang
,
Z.
,
Wang
,
B.
,
Justice
,
N. J.
and
Zheng
,
H.
(
2009
).
Amyloid precursor protein regulates Cav1.2 L-type calcium channel levels and function to influence GABAergic short-term plasticity
.
J. Neurosci
29
,
15660
-
15668
.
Yashiro
,
K.
and
Philpot
,
B. D.
(
2008
).
Regulation of NMDA receptor subunit expression and its implications for LTD, LTP, and metaplasticity
.
Neuropharmacology
55
,
1081
-
1094
.
Yoshida
,
M.
,
Fukuda
,
S.
,
Tozuka
,
Y.
,
Miyamoto
,
Y.
and
Hisatsune
,
T.
(
2004
).
Developmental shift in bidirectional functions of taurine-sensitive chloride channels during cortical circuit formation in postnatal mouse brain
.
J. Neurobiol
60
,
166
-
175
.
Young-Pearse
,
T. L.
,
Ivic
,
L.
,
Kriegstein
,
A. R.
and
Cepko
,
C. L.
(
2006
).
Characterization of mice with targeted deletion of glycine receptor alpha 2
.
Mol. Cell. Biol.
26
,
5728
-
5734
.
Yuryev
,
M.
,
Andriichuk
,
L.
,
Leiwe
,
M.
,
Jokinen
,
V.
,
Carabalona
,
A.
and
Rivera
,
C.
(
2018
).
In vivo two-photon imaging of the embryonic cortex reveals spontaneous ketamine-sensitive calcium activity
.
Sci. Rep.
8
,
16059
.
Yuste
,
R.
,
Peinado
,
A.
and
Katz
,
L. C.
(
1992
).
Neuronal domains in developing neocortex
.
Science
257
,
665
-
669
.
Zacharias
,
D. A.
and
Strehler
,
E. E.
(
1996
).
Change in plasma membrane Ca2(+)-ATPase splice-variant expression in response to a rise in intracellular Ca2+
.
Curr. Biol
6
,
1642
-
1652
.
Zamponi
,
G. W.
,
Striessnig
,
J.
,
Koschak
,
A.
and
Dolphin
,
A. C.
(
2015
).
The physiology, pathology, and pharmacology of voltage-gated calcium channels and their future therapeutic potential
.
Pharmacol. Rev.
67
,
821
-
870
.
Zhang
,
Y.
,
Zhang
,
J.
,
Jiang
,
D.
,
Zhang
,
D.
,
Qian
,
Z.
,
Liu
,
C.
and
Tao
,
J.
(
2012
).
Inhibition of T-type Ca2+ channels by endostatin attenuates human glioblastoma cell proliferation and migration
.
Br. J. Pharmacol.
166
,
1247
-
1260
.
Zhang
,
X.
,
Chen
,
M. H.
,
Wu
,
X.
,
Kodani
,
A.
,
Fan
,
J.
,
Doan
,
R.
,
Ozawa
,
M.
,
Ma
,
J.
,
Yoshida
,
N.
,
Reiter
,
J. F.
et al. 
(
2016
).
Cell-type-specific alternative splicing governs cell fate in the developing cerebral cortex
.
Cell
166
,
1147
-
1162
.

Competing interests

The authors declare no competing or financial interests.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

Supplementary information