Fact-checked by Grok 2 weeks ago

T-type calcium channel

T-type calcium channels, also known as low-voltage-activated (LVA) calcium channels, are a subclass of voltage-gated calcium channels that open in response to small membrane depolarizations near the , typically between -70 and -60 mV, facilitating calcium influx to modulate cellular excitability in both neuronal and non-neuronal tissues. These channels are distinguished by their rapid and inactivation kinetics, small single-channel conductance, and a characteristic "window current" arising from the overlap of their and steady-state inactivation voltage dependencies, which sustains a persistent calcium entry around -60 to -50 mV. Encoded by three genes—CACNA1G (Cav3.1), CACNA1H (Cav3.2), and CACNA1I (Cav3.3)—each consisting of a single pore-forming α1 subunit containing four homologous domains, and lacking the ancillary β and α2-δ subunits common to high-voltage-activated channels. The subtypes differ in their gating properties, pharmacological sensitivities (e.g., Cav3.2 is particularly sensitive to ions), and tissue distribution, with Cav3.2 showing the broadest expression in peripheral tissues. In physiological contexts, T-type channels play pivotal roles in regulating firing patterns, such as generating low-threshold spikes and burst firing in central and peripheral neurons, which contribute to , sleep-wake rhythms, and thalamic oscillations. They also support pacemaker activity in cardiac cells during embryonic development, secretion in neuroendocrine cells, and mechanisms like and depression. Beyond excitable cells, these channels influence non-neuronal functions, including fertilization in , cell proliferation in , and as a second messenger in various tissues. Dysregulation of T-type channels is implicated in several pathologies, underscoring their therapeutic potential; for instance, mutations in CACNA1H (v3.2), including loss-of-function variants linked to disorders and some gain-of-function variants to , while their upregulation contributes to and chemotherapy-induced . In cancer, aberrant expression—particularly of Cav3.1 and v3.2 in , breast, and tumors—promotes , , and to , with studies showing involvement in up to 82% of cases. Selective antagonists targeting these channels have shown promise in preclinical models for treating , , and pain, highlighting their "mixed blessing" as both essential regulators and disease contributors.

Structure

α1 Subunit

The α1 subunit serves as the principal pore-forming component of T-type calcium channels, encoded by genes in the CACNA1G (Cav3.1), CACNA1H (Cav3.2), and CACNA1I (Cav3.3) family. It is synthesized as a single polypeptide chain of approximately 2000 , which folds into a with four homologous (I–IV) connected by intracellular loops. Each consists of six transmembrane segments (S1–S6), conferring a pseudo-tetrameric characteristic of voltage-gated ion channels. The S5 and S6 segments from all four bundle to form the central -conducting pore, while the intervening P-loop regions contribute to the selectivity filter that permits calcium . Voltage sensitivity arises from the S4 segments within each 's voltage-sensing (VSD, comprising S1–S4), which contain a series of positively charged and lysine residues that move in response to changes in , facilitating channel activation. The intracellular loops linking the , particularly those between I–II and II–III, along with the extended C-terminal tail, provide interaction sites for regulatory proteins and post-translational modifications that modulate channel trafficking and function. Across the Cav3 family, the α1 subunit exhibits strong evolutionary conservation in its core topology and key functional motifs, with sequence identities of approximately 60% overall and higher conservation (70-80%) in transmembrane domains among Cav3.1, Cav3.2, and Cav3.3 isoforms, reflecting their shared role in low-voltage-activated calcium influx. High-resolution insights into this architecture have been provided by cryo-EM structures, including those of human Cav3.1 (resolved at 3.1–3.3 Å in 2019) and human Cav3.3 (at 3.3–3.9 Å in 2022), which visualize the full-length α1 subunit embedded in lipid nanodiscs mimicking cellular membranes and reveal conformational details of the VSDs and pore in resting and drug-bound states. Additionally, the cryo-EM structure of human Cav3.2 has been resolved at resolutions ranging from 2.9–3.4 Å in 2024, revealing details of binding sites.

Auxiliary Subunits

T-type calcium channels, primarily composed of the pore-forming α1 subunit from the Cav3 family, can associate with auxiliary β, α2δ, and γ subunits to form multi-subunit complexes that influence channel stability, trafficking to the plasma membrane, and overall expression levels, although these channels remain functional without auxiliaries unlike high-voltage-activated counterparts. These cytoplasmic, extracellular, and transmembrane auxiliaries respectively bind distinct regions of the α1 subunit, promoting proper folding and surface localization while contributing to the formation of the T-type channelosome, a macromolecular that supports synaptic targeting. The β subunits, encoded by four genes (CACNB1–4) and existing as four major isoforms, are intracellular proteins that bind with relatively low affinity to the I–II linker loop of the Cav3 α1 subunit via a conserved α-interaction domain (AID) motif in their guanylate kinase-like domain. This interaction stabilizes nascent channel complexes, protects against endoplasmic reticulum-associated degradation, and enhances membrane trafficking and expression, as demonstrated by co-expression studies where β subunits increase T-type current amplitudes, albeit less dramatically than in high-voltage-activated channels. Experimental evidence from antisense oligonucleotide depletion and isoform-specific co-transfection confirms that β subunits promote assembly of functional T-type channels, with reduced surface expression observed in their absence. The α2δ subunits, comprising four isoforms encoded by CACNA2D1–4, consist of a disulfide-linked extracellular α2 and a transmembrane δ , often glycosyl phosphatidylinositol-anchored. They interact with extracellular loops and sites on the Cav3 α1 subunit, facilitating trafficking through interactions with the and increasing plasma membrane insertion. For T-type channels, co-expression of α2δ-1 or α2δ-2 isoforms approximately doubles Cav3.1 current density by boosting surface expression, as shown in systems. Knockout studies in α2δ-1 mice reveal diminished T-type currents in neurons, underscoring their role in stabilizing channel complexes and supporting physiological localization. The γ subunits, encoded by eight genes (CACNG1–8) and featuring four transmembrane domains, associate with the Cav3 α1 subunit in a tissue-specific manner, particularly in brain regions, to fine-tune channel complexes. Isoforms such as γ2, γ4, and γ5 bind via intracellular loops, contributing to assembly and membrane retention, with co-expression experiments indicating modest enhancements in Cav3.1 trafficking and stability in neuronal contexts. Their involvement is less pronounced than β or α2δ but evident in knockout models where γ absence correlates with altered T-type expression in excitable tissues. The channel complex typically includes one α1 subunit with one β subunit, one α2δ subunit, and optionally one γ subunit, integrating into the channelosome for targeted delivery to synapses. A 2023 review emphasizes how these auxiliaries orchestrate channelosome formation, enabling synaptic enrichment of channels via interactions with trafficking proteins like SCAMP2.

Biophysical Properties

Gating Mechanisms

T-type calcium channels, also known as low-voltage-activated (LVA) channels, exhibit a characteristically low threshold, typically around -60 to -70 mV, which allows them to open in response to small depolarizations from resting membrane potentials. This contrasts sharply with high-voltage-activated (HVA) channels like L-type, which require more depolarized potentials near -20 mV for . The voltage-dependent involves conformational changes in the channel's voltage-sensing domains, primarily driven by the S4 segments, enabling T-type channels to contribute to subthreshold excitability. The gating kinetics of T-type channels are marked by rapid activation and inactivation, distinguishing them from other calcium channel types. Activation time constants (τ_act) range from approximately 1-7 ms across isoforms (Ca_v3.1, Ca_v3.2, Ca_v3.3), accelerating with stronger depolarizations, while inactivation time constants (τ_inact) vary from 11-69 ms, with Ca_v3.3 showing the slowest rates. Recovery from inactivation occurs over hundreds of milliseconds at hyperpolarized potentials, such as τ_r ≈ 270 ms at -80 mV, allowing channels to reset following transient openings. These fast kinetics result in transient currents that peak and decay quickly, typically within tens of milliseconds at physiological temperatures. A key feature of T-type gating is the window current, arising from the overlap of steady-state and inactivation curves around -60 to -50 mV, where a small fraction of channels (<1%) remains open, permitting sustained calcium influx at subthreshold potentials without full depolarization. This non-inactivating component supports tonic excitability and is particularly prominent in isoforms like Ca_v3.1. Gating also displays hysteresis, where prior hyperpolarization shifts the curve to more negative voltages due to enhanced recovery from inactivation, effectively lowering the threshold for subsequent activations compared to depolarized histories. Such path-dependent behavior underscores the channels' role in burst firing patterns. Mathematical modeling of T-type currents often employs Hodgkin-Huxley-style formulations to capture these dynamics, with the current expressed as I_T = g_T m^3 h (V - E_{Ca}), where g_T is the maximal conductance, m represents voltage-dependent activation (with three identical gates), h denotes inactivation, V is membrane potential, and E_{Ca} is the calcium reversal potential. Rate constants for m and h are derived from voltage-clamp data, incorporating voltage-dependent transitions (e.g., α_m and β_m for activation) to simulate the transient nature and low-threshold spike generation. In experimental settings, patch-clamp recordings distinguish T-type currents by their activation at low voltages (~ -60 mV), rapid inactivation (transient profile), and slow deactivation tails, contrasting with the sustained currents of HVA channels like . Whole-cell recordings in neurons, for instance, reveal peak T-currents at -40 mV with monophasic decay, while single-channel analysis shows small conductances (7-12 pS) and brief openings, confirming the biophysical signature in native tissues.

Ionic Selectivity and Permeation

T-type calcium channels exhibit high selectivity for Ca²⁺ ions over monovalent cations such as Na⁺ and K⁺, primarily due to the EEDD locus in the selectivity filter formed by glutamate residues in domains I and II and aspartate residues in domains III and IV within the S5–S6 linkers of the α1 subunit. This negatively charged ring coordinates divalent cations with high affinity, enabling Ca²⁺ to bind tightly and exclude monovalent ions through electrostatic repulsion and dehydration barriers in the pore. The selectivity ratio for Ca²⁺ over Na⁺ is approximately 20-70:1 under physiological conditions, varying by isoform (e.g., Ca_v3.1 ≈68:1, Ca_v3.2 ≈47:1, Ca_v3.3 ≈21:1). The single-channel conductance of T-type channels for Ca²⁺ is approximately 1 pS in physiological solutions (∼1–2 mM Ca²⁺), reflecting a narrow pore diameter that limits ion flux compared to high-voltage-activated channels. Permeation occurs via a multi-ion mechanism involving multiple occupancy of the selectivity filter, where incoming Ca²⁺ ions facilitate a "knock-off" process to dislodge resident ions and prevent anomalous block at micromolar Ca²⁺ levels. This cooperative ion-ion interaction supports rapid throughput, with fractional Ca²⁺ currents saturating at low millimolar concentrations due to surface charge effects and reduced block. T-type channels also permit permeation of other divalent cations, with relative permeability approximately equal for Ba²⁺, Sr²⁺, and Ca²⁺, allowing these ions to serve as charge carriers in experimental settings. Pharmacologically, they are distinguished by sensitivity to block by Cd²⁺ and Ni²⁺, with IC₅₀ values in the micromolar range, serving as hallmarks for isolating T-type currents from other Ca²⁺ channel types. Current-voltage relations display peak inward currents between -40 and -20 mV, reflecting the low-voltage activation range and reversal potential near the Ca²⁺ equilibrium. Recent cryo-EM structures of T-type channels, such as Ca_v3.2 and Ca_v3.3, reveal a hydrated entry pathway into the pore, where Ca²⁺ ions approach the selectivity filter in a partially hydrated state before coordination by the EEDD ring, facilitating selective dehydration and binding. More recent 2024 structures of Ca_v3.2 in complex with selective antagonists further elucidate inhibition mechanisms at the selectivity filter and activation gate. These models highlight the role of the inner pore vestibule in stabilizing multi-ion configurations, consistent with the knock-off permeation dynamics observed electrophysiologically.

Regulation

Post-Translational Modifications

Post-translational modifications play a critical role in regulating the activity, trafficking, and stability of T-type calcium channels, primarily through enzymatic alterations to the α1 subunits (Cav3 family). Phosphorylation by kinases such as and targets intracellular loops, shifting the voltage dependence of activation to more negative potentials for certain kinases like PKC, while others like PKA may enhance availability without altering voltage dependence. For instance, PKC phosphorylation at serine/threonine sites on Cav3.2 slows inactivation kinetics, thereby amplifying calcium influx during low-threshold activation. Dephosphorylation by protein phosphatases, including calcineurin, counteracts these effects by promoting channel inactivation and reducing surface expression, maintaining a dynamic balance in channel responsiveness. The interplay between kinases and phosphatases is highlighted in a 2023 review, which details site-specific modulation across Cav3.1, Cav3.2, and Cav3.3 isoforms, emphasizing how phosphatase activity fine-tunes gating under physiological conditions. Glycosylation, particularly N-linked modifications on extracellular loops, influences channel trafficking and surface expression, with deglycosylation mutants exhibiting reduced peak currents and altered gating properties in . These modifications stabilize the channel in the plasma membrane and modulate permeability to ions like zinc, impacting neuronal excitability. Ubiquitination targets T-type channels for proteasomal degradation, reducing surface levels in sensory neurons, while SUMOylation regulates interactions with deubiquitinating enzymes like USP5, thereby controlling channel stability and trafficking. Experimental evidence from phosphomimetic mutants (e.g., serine-to-aspartate substitutions on ) demonstrates that mimicking phosphorylation shifts activation curves to more negative potentials, confirming the direct impact on gating without requiring kinase activity.

Protein Interactions

T-type calcium channels (Cav3 family) form part of a multiprotein termed the "T-type channelosome," which encompasses non-covalent interactions with various binding partners that govern channel localization, trafficking, and functional modulation in diverse cellular contexts. This channelosome enables precise control over channel expression at the plasma membrane, influencing physiological processes such as neuronal excitability and cardiac rhythmicity. Seminal reviews highlight how these macromolecular assemblies integrate channels into signaling networks, with interactors binding to intracellular domains to stabilize or regulate activity. Key components of the channelosome include SNARE proteins syntaxin-1A and SNAP-25, which interact directly with the C-terminal domain of Cav3.2 to facilitate synaptic targeting and low-threshold in neurons. These interactions enhance channel clustering at presynaptic sites, promoting calcium influx that supports release near resting potentials. Cytoskeletal elements, such as B and spectrin (α/β), bind to T-type channels to anchor them to the membrane , ensuring stability and proper localization in both neuronal and cardiac tissues; disruption of these links alters channel surface expression and . G-protein βγ subunits exert inhibitory effects on channels through direct binding to the intracellular II-III loop domain, particularly of Cav3.2, leading to a voltage-independent reduction in channel activity that fine-tunes excitability in response to G-protein-coupled receptor signaling. In neuronal dendrites, scaffolds like CACHD1 (an α2δ-like protein) and SCAMP2 regulate Cav3 localization and surface expression, supporting dendritic of synaptic inputs. Tissue-specific complexes further diversify ; for instance, in cells, binds to Cav3 channels to mediate calcium-dependent modulation of gating, influencing without relying on traditional IQ motifs. Recent proteomic approaches, including co-immunoprecipitation coupled with , have expanded the channelosome by identifying novel interactors that link T-type channels to broader cellular pathways, such as trafficking and signaling scaffolds. These studies underscore the dynamic nature of the complex, revealing partners that adapt channel function to specific physiological demands across tissues.

Physiological Functions

Neuronal Excitability

T-type calcium channels play a pivotal role in modulating neuronal excitability by facilitating burst firing and low-threshold spikes (LTS) in relay neurons. These channels, activated at relatively hyperpolarized potentials following deinactivation during membrane hyperpolarization, generate regenerative LTS that trigger bursts of action potentials, thereby enhancing the responsiveness of relay neurons to sensory inputs. This burst mode contrasts with tonic firing and is essential for rhythmic network activity, as demonstrated in electrophysiological studies where blockade of T-type currents abolishes LTS and reduces burst propensity. A hallmark electrophysiological signature is rebound excitation, where hyperpolarization deinactivates T-type channels, leading to a post-hyperpolarization and subsequent burst firing upon return to ; this mechanism has been observed across various central neurons, including those in the and inferior olive. In the thalamocortical circuit, T-type channels contribute to the generation of sleep spindles and absence seizures by promoting synchronized burst firing between relay neurons and reticular thalamic nuclei. The LTS-mediated bursts facilitate oscillatory rhythms at 7-14 Hz characteristic of sleep spindles, while excessive bursting can lead to the pathological 3 Hz spike-and-wave discharges seen in absence seizures. Beyond the thalamus, T-type channels support dendritic Ca²⁺ spikes in hippocampal and cortical pyramidal neurons, which amplify synaptic inputs and drive burst firing to influence synaptic plasticity. For instance, in CA1 hippocampal neurons, these dendritic spikes, often involving Cav3.1 and Cav3.2 isoforms, enable spike-timing-dependent plasticity by providing localized Ca²⁺ influx necessary for long-term potentiation induction. Similarly, in layer 5 cortical neurons, T-type-mediated dendritic bursts facilitate Hebbian plasticity rules, linking pre- and postsynaptic activity to strengthen specific synapses. T-type channels also regulate post-synaptic excitability in sensory neurons, particularly in dorsal root ganglia, where they lower the threshold for initiation and modulate input-output relationships. In these neurons, T-type currents enhance excitability by contributing to afterdepolarizations following inhibitory inputs, thereby fine-tuning sensory . Expression of T-type channels is developmentally regulated, with high levels in immature neurons supporting early network formation and excitability; for example, Cav3.2 channels are abundant in embryonic and early postnatal stages, promoting neuritogenesis and synaptic maturation, but decline in adulthood as neurons shift toward firing patterns. Recent advances highlight T-type channels' role in presynaptic modulation of release, particularly in terminals where Cav3 isoforms contribute to Ca²⁺-dependent vesicle alongside high-voltage-activated channels, influencing reward-related signaling and potentially synaptic efficacy.

Cardiac Pacemaking

T-type calcium channels play a critical role in the () by contributing to in cells through their window currents, which occur at the overlap of activation and inactivation voltage ranges around -50 to -40 mV. These low-voltage-activated currents provide a sustained inward Ca²⁺ flux during phase 4 of the action potential, facilitating the gradual that leads to the next spontaneous firing. In cells, this mechanism amplifies the initial initiated by other currents, such as the funny current (I_f), ensuring reliable pacemaking. T-type channels are co-expressed with hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in cells, where they synergistically support phase 4 . HCN channels, primarily HCN4 in the , generate the initial slow via I_f, while T-type channels (particularly Cav3.1) activate subsequently to accelerate the process toward , enhancing overall . This interplay is evident in atrioventricular node cells, where co-expression promotes robust phase 4 slopes. Disruption of this coordination, as seen in HCN4 mutations, can impair T-type contributions and lead to . Species differences in T-type channel prominence are notable, with higher expression and functional impact in atria compared to tissue. In mice, Cav3.1 channels are abundant in the and support significant pacemaking, as reduces heart rate by approximately 10-30%. In contrast, atrial and tissues show lower T-type densities, with contributions more evident in pathological states like rather than baseline . Rabbit models further highlight this gradient, showing negligible T-type involvement in peripheral regions. Recent studies using models have demonstrated that channel ablation reduces in arrhythmia-prone hearts. In mouse models of , Cav3 impairs escape rhythms and increases lethal incidence by diminishing subsidiary pacemaker activity. A 2023 analysis in genetic models confirmed that loss exacerbates and conduction defects, underscoring their protective role against excessive rhythm slowing. Sympathetic modulation enhances currents via β-adrenergic stimulation, which increases channel activity through phosphorylation, thereby accelerating . This regulation is vital for fight-or-flight responses, as isoproterenol application boosts currents and shifts to more negative potentials in isolated cardiomyocytes.

Roles in Non-Excitable Tissues

calcium channels contribute to diverse physiological processes in non-excitable tissues by enabling low-threshold Ca²⁺ influx that supports localized signaling, often independent of action potentials. In secretory and contractile cells, these channels facilitate Ca²⁺ oscillations and events critical for release and mechanical responses. Their expression in reproductive, skeletal, renal, and cells underscores their role in , motility, and structural maintenance, with dysregulation linked to impaired cellular function. In pancreatic β-cells, calcium channels, particularly the Caᵥ3.2 isoform, mediate glucose-stimulated insulin through the generation of Ca²⁺ oscillations and burst-like electrical activity that couples to . These channels lower the activation threshold for Ca²⁺ entry, enhancing excitability during low-glucose conditions and contributing up to 60-70% of insulin release at 6 mM glucose in human β-cells. Pharmacological inhibition or genetic knockdown of Caᵥ3.2 impairs this process, highlighting their necessity for normal β-cell function and glucose . Within vascular smooth muscle cells, T-type channels regulate contraction and myogenic tone by providing transient Ca²⁺ currents that amplify and sustain intracellular Ca²⁺ levels for actin-myosin interactions. Expressed alongside L-type channels, they support pressure-induced in resistance arteries, such as cremaster arterioles, where selective blockade reduces tone without affecting relaxation. Additionally, these channels influence proliferation in vascular remodeling, linking Ca²⁺ signaling to responses in the vessel wall. In reproductive cells, T-type channels drive sperm motility and the acrosome reaction, an exocytotic event essential for fertilization triggered by zona pellucida binding. These channels activate at low voltages to initiate Ca²⁺-dependent flagellar beating and acrosomal vesicle fusion, with mibefradil-sensitive currents observed in mammalian sperm. T-type channels also participate in bone remodeling via osteoclast differentiation and resorption. The Caᵥ3.2 isoform responds to RANKL signaling to generate Ca²⁺ transients that promote precursor fusion and podosome organization in osteoclasts, facilitating matrix degradation. RNA interference-mediated knockdown of Caᵥ3.2 in bone marrow-derived macrophages significantly suppresses osteoclastogenesis, reducing bone resorption markers and preserving bone mass. Non-canonical functions of channels extend to s, where they influence and through PDGF-stimulated Ca²⁺ signaling. In these cells, T-type-mediated Ca²⁺ entry sustains growth factor-induced transients that activate downstream pathways like MAPK, promoting and tissue repair without inducing . Blockade with mibefradil inhibits fibroblast proliferation in response to mitogens, underscoring their role in non-malignant stromal dynamics.

Isoforms and Genetic Variation

Cav3.1

The Cav3.1 isoform of the T-type calcium channel is encoded by the , located on human chromosome 17q21.33. This gene undergoes extensive , with at least six exons showing variability, including those in the C-terminal region that influence channel trafficking, modulation, and biophysical properties. For instance, C-terminal splice variants can alter interactions with regulatory proteins and affect the channel's voltage dependence and inactivation rates. Cav3.1 is predominantly expressed in the , particularly in the , , and subthalamic nucleus, with moderate levels in the heart and lower expression in other tissues like and . In the , it contributes to neuronal excitability in relay and reticular thalamic neurons, while in cardiac tissue, it supports pacemaker activity during development, becoming the dominant isoform in adult cardiomyocytes. Biophysically, Cav3.1 activates at low voltages around -60 , with inactivation kinetics that are faster than those of Cav3.3 but similar to or slightly slower than Cav3.2, enabling sustained low-threshold currents. It is distinguished by prominent rebound currents following hyperpolarizing pulses, which arise from its relatively rapid recovery from inactivation ( ~100-200 ms) compared to other isoforms, facilitating burst firing in thalamic neurons. This property underpins its role in generating rhythmic oscillations. In the , Cav3.1 plays a key role in pacemaking for non-rapid eye movement (NREM) sleep rhythms, where it mediates low-threshold spikes that drive burst firing in thalamocortical circuits, contributing to delta oscillations (1-4 Hz) and sleep spindles. Knockout studies in mice show reduced low-frequency thalamocortical oscillations and disrupted sleep architecture, highlighting its essential contribution to these physiological rhythms. Mutations in CACNA1G have been linked to gain-of-function effects in (IGE), with discoveries in the early identifying variants like p.Ala570Val that may increase neuronal excitability. These mutations enhance thalamic burst firing, promoting absence seizures characteristic of IGE. Genetic screening identifies CACNA1G as a strong candidate risk gene. As of , recent studies highlight functional expression of channels in sensory neurons, with implications for isoform-specific genetic variations in neurodevelopmental contexts.

Cav3.2

The Cav3.2 channel, encoded by the CACNA1H located on chromosome 16p13.3, is a low-voltage-activated calcium that plays a critical role in neuronal excitability, particularly in sensory pathways. This undergoes extensive at 12–14 sites, generating over 4,000 potential mRNA transcripts, with at least eight functional variants that exhibit distinct gating properties when expressed in mammalian cells. These variants influence (half-activation voltages ranging from -39 to -44 mV), steady-state inactivation (midpoints spanning over 10 mV), and , leading to variations in currents up to 12-fold and altered /inactivation time constants that can differ by threefold. Cav3.2 demonstrates high expression in the dorsal root ganglia (DRG), where approximately 20% of neurons—predominantly small- and medium-sized (20–30 μm diameter)—express its mRNA and protein, contributing to nociceptive and mechanoreceptive functions. In peripheral nerves, it localizes to endings and mechanoreceptors such as lanceolate afferents, but is absent from slowly adapting mechanoreceptors like Merkel cell-associated endings. Within the , expression is notable in select brain regions, including the of the , , and , where it supports burst firing in neurons involved in . Distinct biophysical properties distinguish Cav3.2 from other T-type isoforms, including high sensitivity to block at low micromolar concentrations (selective inhibition at 10–50 μM), which targets a residue in the channel's extracellular loop. It also features rapid inactivation kinetics, with time constants for inactivation faster than those of high-voltage-activated channels, enabling transient calcium influx that promotes low-threshold . Additionally, Cav3.2 is modulated by (H₂S), a gasotransmitter produced by cystathionine-γ-lyase, which exerts concentration-dependent effects: inhibition at low levels (10 μM) via modification of extracellular cysteines and enhancement at higher levels (100 μM) through interaction at a zinc-binding site involving His191, thereby amplifying channel activity in pathological states. In sensory pathways, Cav3.2 contributes to peripheral and by enhancing neuronal excitability during . Following carrageenan-induced paw in mice, Cav3.2 mRNA and protein levels increase 2.1-fold in ipsilateral DRG neurons within 1–2 days, with pronounced upregulation (twofold) in TRPV1-positive nociceptors, correlating with heightened and . Pharmacological blockade with T-type antagonists like mibefradil or NNC 55-0396 attenuates this , confirming Cav3.2's necessity for inflammatory maintenance, as its silencing via intrathecal delivery reduces behavioral responses to noxious stimuli. Missense mutations in CACNA1H have been identified in individuals with , occurring in approximately 1.3% of cases (6 out of 461 screened), and functional analyses reveal altered gating properties consistent with loss-of-function effects that disrupt in neuronal networks. These variants, often or inherited, impair channel activity and contribute to ASD phenotypes by affecting synaptic development and excitability in brain regions like the and . Recent advances highlight Cav3.2's upregulation in neuropathic pain through inflammatory mechanisms, where peripheral nerve injury or inflammation increases channel expression in DRG via enhanced interactions with deubiquitinating enzymes like USP5, sustaining hyperexcitability and allodynia. A 2023 review emphasizes the CSE/H₂S/Cav3.2 pathway's role, showing that H₂S-driven augmentation of Cav3.2 currents in DRG neurons post-injury exacerbates neuropathic symptoms, with genetic knockout abolishing H₂S-dependent pain behaviors in rodents and positioning this axis as a target for novel analgesics. As of 2025, studies report sex dimorphism in Cav3.2 functional expression in DRG neurons and its role in shaping firing patterns during maturation.;

Cav3.3

The , encoded by the , is located on 22q13.1 spanning approximately 119 kb. Unlike other isoforms, CACNA1I produces fewer variants, with five principal transcripts identified in , contributing to limited molecular diversity compared to CACNA1G or CACNA1H. Cav3.3 is prominently expressed in the (nRt), where it supports burst firing essential for thalamocortical oscillations. It is also present in pituitary cells, including somatotrophs and gonadotrophs, facilitating calcium influx that regulates hormone secretion. Expression extends to neurons involved in respiratory control, contributing to rhythm generation in networks like the . Biophysically, Cav3.3 displays the slowest activation and inactivation kinetics among T-type channels, with ultra-slow inactivation time constants exceeding those of Cav3.1 by an , enabling prolonged low-threshold bursts. Its activation threshold is the most negative, around -65 to -70 , allowing operation near resting potentials to fine-tune excitability. These properties underpin roles in via thalamic circuits, where Cav3.3 modulates oscillatory activity in the nRt to influence cerebellar and inputs for coordinated movement. In the pituitary, it drives depolarization-induced calcium entry critical for (GH) and follicle-stimulating hormone (FSH) release, with blockade reducing secretory bursts in somatotrophs. Rare loss-of-function mutations in CACNA1I disrupt channel gating, leading to reduced and impaired burst firing, and have been associated with through altered thalamic spindle activity. Gain-of-function variants, identified in patients with disorder and , shift activation to more hyperpolarized potentials, exacerbating neuronal hyperexcitability. Recent analyses confirm convergence of CACNA1I variants across and neurodevelopmental disorders, highlighting shared disruptions in calcium-dependent .

Disease Associations

Epilepsy and Seizures

T-type calcium channels, particularly the Cav3.2 isoform, play a critical role in the of through genetic and acquired dysregulation that enhances neuronal burst firing and network synchrony. Gain-of-function and polymorphisms in CACNA1H (encoding Cav3.2) have been associated with increased activity, leading to hyperexcitability in thalamocortical circuits and contributing to childhood absence (CAE). For instance, certain missense variants in CACNA1H, such as R212C, result in altered gating properties that prolong opening, thereby amplifying calcium influx, though primarily linked to disorders with comorbidities. These genetic alterations underscore channels as susceptibility factors in idiopathic generalized epilepsies, where even subtle shifts in kinetics can disrupt normal thalamic relay function. In the , T-type channels mediate low-threshold spikes that underlie rebound burst firing in relay and reticular neurons, a mechanism that amplifies oscillatory activity and generates the characteristic 3-Hz spike-wave discharges (SWDs) observed in absence epilepsy on EEG. Hyperactive Cav3.2 channels in thalamic neurons facilitate prolonged burst durations and synchronized discharges across thalamocortical loops, directly contributing to the initiation and propagation of SWDs. This burst amplification is particularly evident in models where T-type is increased, leading to pathological rhythmicity that correlates with onset. Acquired upregulation of channels also contributes to epileptogenesis in (TLE), where prolonged seizures induce transcriptional changes that enhance channel expression and function. In rodent models of TLE induced by , Cav3.2 mRNA and protein levels are transiently upregulated in hippocampal CA1 pyramidal neurons, resulting in increased dendritic T-type currents that promote burst firing and long-term hyperexcitability. This activity-dependent persists for weeks post-insult, fostering a pro-epileptic state in the and contributing to spontaneous recurrent seizures in TLE models. Animal studies using Cav3 knockout models have demonstrated reduced seizure susceptibility, highlighting the pro-convulsant role of T-type channels. Cav3.1 knockout mice exhibit diminished thalamic burst firing and resistance to pharmacologically induced absence seizures, such as those triggered by baclofen or γ-hydroxybutyrate, with fewer and shorter SWDs compared to wild-type controls. Similarly, Cav3.2 knockout or knockdown in genetic absence epilepsy rats (GAERS) reduces spike frequency in reticular thalamic bursts and shortens seizure duration, confirming that T-type hyperactivity sustains epileptic networks from the 2000s through recent 2023 analyses. T-type channel inhibition is explored in (DS), a developmental primarily caused by SCN1A loss-of-function mutations. inhibits Cav3.1/3.2/3.3 channels to suppress thalamocortical oscillations and reduce frequency in DS patients. Diagnostically, EEG patterns in absence , such as SWDs, reflect underlying T-type-mediated rebound potentials in thalamic neurons, where post-hyperpolarization bursts generate the spike component of discharges. These correlates aid in identifying T-type dysregulation, as enhanced rebound excitability on intracellular recordings aligns with the 2-4 Hz rhythmicity of clinical SWDs, supporting targeted genetic screening for channelopathies.

Chronic Pain Conditions

T-type calcium channels, particularly the Cav3.2 isoform, play a significant role in the pathophysiology of chronic pain conditions such as neuropathic and inflammatory pain. Following peripheral nerve injury, such as spinal nerve ligation, there is a marked upregulation of Cav3.2 expression and function in dorsal root ganglion (DRG) neurons, including both damaged and adjacent intact neurons. This increase enhances low-threshold calcium influx, promoting neuronal hyperexcitability and contributing to the generation of ectopic firing in primary sensory afferents. In the spinal cord, upregulated T-type channels in dorsal horn neurons facilitate burst firing and synaptic amplification, which underpin central wind-up—a key mechanism of central sensitization where repeated nociceptive inputs lead to exaggerated pain responses. These peripheral and central changes collectively drive mechanical allodynia and hyperalgesia in rodent models of neuropathic pain. In painful (PDN), induces elevated expression of Cav3.2 in small-diameter DRG neurons, leading to enhanced currents and hyperexcitability. This upregulation is mediated by post-translational modifications like , which sensitize the channels and contribute to thermal and mechanical in streptozotocin-induced diabetic rats. Early PDN is characterized by a selective increase in in capsaicin-sensitive nociceptors, correlating with spontaneous ectopic activity and hypersensitivity. Endogenous modulators such as (H2S) and further influence Cav3.2 function in pain states. H2S, produced by cystathionine-γ-lyase in activated and macrophages post-injury, enhances Cav3.2 currents in a concentration-dependent manner (facilitation at 100 µM), promoting sensitization and maintenance of . typically inhibits Cav3.2 at a high-affinity site involving 191, but changes induced by H2S or reducing agents relieve this inhibition, thereby enhancing channel activity and contributing to in models like L5 ligation and paclitaxel-induced neuropathy. Preclinical studies demonstrate the efficacy of selective channel blockers in alleviating symptoms. Compounds like Z944 and ABT-639 reduce T-type currents in DRG and spinal neurons, attenuating mechanical and thermal in rat models of and inflammatory pain without affecting normal sensory thresholds. These blockers interrupt ectopic firing and central wind-up, highlighting T-type channels as viable targets. In humans, genetic variants in CACNA1H, which encodes Cav3.2, are associated with altered channel function and susceptibility to with aura—a disorder involving and sensory hypersensitivity. Whole-exome sequencing of patients reveals an increased burden of missense variants in CACNA1H, acting as genetic modifiers that enhance T-type channel activity and pain propagation.

Cardiovascular and Metabolic Disorders

T-type calcium channels, particularly Cav3.1 and Cav3.2 isoforms, are expressed in the where they contribute to cardiac pacemaking and . Dysregulation of these channels has been implicated in . Concomitant ablation of Cav1.3 and Cav3.1 disrupts function, leading to and arrhythmias through impaired impulse generation. In vascular smooth muscle, calcium channels mediate low-voltage-activated currents that promote by facilitating calcium entry at depolarized potentials near the resting membrane voltage. channels contribute to myogenic tone and contractility in resistance arteries. Blockade of these channels attenuates vasoconstrictor responses in preclinical models. calcium channels also play a key role in aldosterone secretion from adrenal cells, where they support calcium oscillations triggered by angiotensin II and , essential for steroidogenesis. Both and L-type channels equally regulate this process, with channels enabling low-threshold calcium influx that sustains secretory bursts. Aldosterone can upregulate channel expression in glomerulosa cells via , potentially amplifying production in response to stimuli like high sodium intake, promoting renal sodium retention and elevation via mineralocorticoid receptor activation. A 2023 meta-analysis of randomized controlled trials compared N-/T-type calcium channel blockers with L-type blockers in patients with on renin-angiotensin system inhibitors, finding that N-/T-type agents more effectively reduced and without adversely affecting or , unlike some L-type blockers that may dilate and worsen intraglomerular pressure. This suggests superior renoprotective effects of T-type blockade in preserving kidney function during hypertensive nephropathy, including . In metabolic disorders, calcium channels, especially Cav3.2, regulate insulin secretion in pancreatic β-cells by enhancing excitability and coupling glucose-stimulated to calcium-dependent . Dysfunction or downregulation of these channels in impairs glucose-induced insulin release, contributing to β-cell failure and through reduced second-phase secretion and altered electrical bursting patterns. This channel impairment, often linked to and , represents a key mechanism in β-cell exhaustion during disease progression.

Cancer and Neurodegeneration

T-type calcium channels, particularly the Cav3.2 isoform, have been implicated in oncogenic processes across several cancer types through dysregulation of Ca²⁺ signaling that drives and . In , Cav3.2 is overexpressed in cells during neuroendocrine differentiation induced by androgen depletion or inflammatory cytokines, leading to enhanced Ca²⁺-dependent secretion of prostate acidic phosphatase and promotion of ; siRNA-mediated knockdown of Cav3.2 reduces in these cells. Similarly, pharmacological inhibition of Cav3.2 with mibefradil decreases cell viability and neurite outgrowth—a proxy for migratory behavior—in neuroendocrine-differentiated models. In , Cav3.2 mRNA levels are elevated in trastuzumab-resistant SKBR3 cells and correlate with poorer prognosis in estrogen receptor-positive subtypes; overexpression of Cav3.2 in cells upregulates mRNA markers associated with (e.g., , MMP9) and (e.g., ), mediated by altered Ca²⁺ influx and signaling pathways. Inhibition of T-type channels with mibefradil or ML218 suppresses in cells, highlighting their pro-tumorigenic role via sustained Ca²⁺ elevation. In gliomas, T-type calcium channels contribute to tumor invasion and , with Cav3.2 prominently expressed in stem-like cells (GSCs) and correlating with poor patient survival. Blockade of T-type channels with mibefradil inhibits GSC proliferation, induces , and reduces invasion in U87 glioma cells by disrupting Ca²⁺ homeostasis and downregulating pro-angiogenic factors such as PDGFA, PDGFB, and TGFB1. shRNA knockdown of Cav3.2 mimics these effects, suppressing tumor growth in orthotopic xenograft models and sensitizing cells to chemotherapy. Experimental siRNA knockdown of Cav3.2 in glioma xenografts further confirms inhibition of tumor progression, reducing overall growth and metastatic potential through impaired Ca²⁺-driven motility. In neurodegeneration, T-type channel hyperactivity contributes to neuronal dysfunction in (), particularly in the where compensatory upregulation of Cav3 channels in neurons exacerbates vulnerability to stressors like . This hyperactivity promotes burst firing and aberrant Ca²⁺ influx, contributing to in models; T-type blockers like mibefradil reduce burst discharges in neurons, alleviating motor symptoms. T-type channels exhibit a dual role in , with blockade exerting neuroprotective effects by mitigating Ca²⁺ overload that promotes α-synuclein aggregation; in cellular models, T-type inhibition with trimethadione prevents Ca²⁺-dependent α-synuclein fibrillization and reduces aggregate formation. T-type channel blockers have shown neuroprotective effects in amyotrophic lateral sclerosis (ALS) models by reducing motor neuron excitotoxicity. Pimozide protects vulnerable motor neurons by normalizing Ca²⁺ signaling in TDP-43 transgenic models.

Pharmacological Targeting

Known Modulators

T-type calcium channels are modulated by a variety of pharmacological agents, including inorganic ions, organic compounds, and natural substances, which primarily act as antagonists, with limited agonists identified. These modulators target the channel's pore or gating mechanisms, influencing low-voltage-activated currents in excitable cells. Inorganic blockers such as nickel ions (Ni²⁺) exhibit selectivity for T-type channels at low micromolar concentrations, with an IC₅₀ of approximately 13 μM for Caᵥ3.2, while higher concentrations (above 50 μM) are required to block high-voltage-activated channels like L-type, enabling their use to isolate T-type currents in electrophysiological studies. Mibefradil, another inorganic-like blocker, inhibits T-type channels with IC₅₀ values in the 1-5 μM range but shows only 10- to 20-fold selectivity over L-type channels, limiting its specificity; it was developed as a cardiovascular agent but voluntarily withdrawn from the market in 1998 due to severe drug interactions leading to risks like rhabdomyolysis and cardiac arrhythmias. Organic antagonists developed in the offer improved selectivity and state-dependent blockade, particularly for the Caᵥ3.2 isoform implicated in signaling. Z944, a piperidine-based compound, potently blocks all three isoforms (Caᵥ3.1, Caᵥ3.2, Caᵥ3.3) with IC₅₀ values of 50-160 nM, demonstrating over 100-fold selectivity against L-type and other voltage-gated channels, and binds within the channel's intracellular gate to stabilize the closed state. TTA-P2, a derivative, acts as a state-dependent blocker preferentially inhibiting inactivated or open states of Caᵥ3.2 with an IC₅₀ of about 20 nM, showing minimal effects on high-voltage-activated channels and reducing neuronal hyperexcitability in models without altering baseline activity. These compounds highlight advances in isoform-specific targeting, though challenges persist in achieving absolute selectivity across T-type subtypes and avoiding off-target effects on sodium or potassium channels. Agonists for T-type channels remain limited, with zinc ions (Zn²⁺) providing one example of enhancement at concentrations relevant to physiological conditions. At low micromolar levels (e.g., 1-10 μM total Zn²⁺, with free levels around 100 nM in synaptic clefts), Zn²⁺ potentiates Caᵥ3.3 currents by slowing deactivation , increasing calcium influx during repetitive firing, while it inhibits Caᵥ3.1 and Caᵥ3.2 at similar or higher concentrations, underscoring subunit-specific in neuronal signaling. Natural modulators include (H₂S) donors, which exert concentration-dependent effects on Caᵥ3.2. At higher concentrations (e.g., 100 μM Na₂S or 1.5 mM NaHS), H₂S donors potentiate Caᵥ3.2 currents by altering redox-sensitive sites like His191 and extracellular cysteines, enhancing channel activity in sensory neurons; lower concentrations (10 μM) instead inhibit, contributing to dual roles in pain modulation as detailed in recent reviews. Anesthetics such as also inhibit T-type channels, contributing to their effects. reduces Caᵥ3.1 and Caᵥ3.2 currents by shifting the voltage dependence of activation toward more depolarized potentials (gating shift) and modulating intracellular PIP₂ signaling, with effects observed at clinically relevant concentrations (10-50 μM). Selectivity remains a key challenge for T-type modulators, as many compounds exhibit varying affinities across isoforms and overlap with other types, necessitating careful IC₅₀ profiling. For instance, while Ni²⁺ and Z944 provide good discrimination (IC₅₀ ratios >10-100 for T- vs. L-type), mibefradil's lower selectivity (10-20-fold) led to off-target cardiovascular effects, emphasizing the need for structure-based design to minimize interactions with high-voltage-activated channels in therapeutic development.

Therapeutic Applications

T-type calcium channels have emerged as promising therapeutic targets in several clinical contexts, particularly where their modulation can alleviate pathological hyperexcitability or cellular proliferation. In the management of chronic pain, such as painful diabetic neuropathy (PDN), the selective T-type channel modulator Z944 has demonstrated efficacy in preclinical models of neuropathic and inflammatory pain by reducing neuronal excitability without significant motor impairment. A Phase Ib clinical trial completed in 2014 assessed safety in healthy volunteers and patients with neuropathic pain, but no further clinical advancement has been reported as of 2025. For , remains a first-line for absence seizures, primarily due to its non-selective blockade of channels, which suppresses thalamocortical oscillations underlying these episodes. Clinical guidelines endorse its use in children and adults with newly diagnosed absence , showing seizure freedom rates of approximately 70% in responsive patients, though it lacks broad efficacy against other . In cardiovascular applications, , a partial and blocker, is employed for treating vertigo associated with disorders and as an adjunct in management by improving vestibular function and reducing vascular contraction. Its inhibitory effects contribute to symptom relief in conditions like Meniere's disease, with clinical evidence supporting reduced vertigo episodes in over 80% of patients when combined with . Therapeutic potential extends to , where preclinical studies of T-type antagonists, such as KTt-45, have shown inhibition of and by disrupting calcium-dependent signaling pathways in models of , , and cancers. In , T-type channel blockers have alleviated levodopa-induced in preclinical rodent models by modulating subthalamic nucleus burst firing, thereby restoring motor balance without exacerbating parkinsonian symptoms. Local application of these agents reduced severity by up to 50% in 6-hydroxydopamine-lesioned rats, highlighting their potential as adjunct therapies to levodopa. Despite these advances, therapeutic targeting of channels faces challenges, including off-target effects on other ion channels that can lead to or unintended neuronal silencing. Reviews on and highlight the potential of T-type inhibition for mitigating ischemic brain injury during surgical procedures, but emphasize the need for isoform-selective modulators to minimize systemic side effects like .

References

  1. [1]
    Neuronal T–type calcium channels: What's new? Iftinca
    This review summarizes recent advances in our understanding of neuronal T–type calcium channel regulation as well as their physiological and pathophysiological ...
  2. [2]
    T-Type Calcium Channels: A Mixed Blessing - PMC - PubMed Central
    Aug 31, 2022 · This review puts into context the relevance of T-type calcium channels in cancer and in chemotherapy side effects, considering also the cardiotoxicity.
  3. [3]
    Molecular physiology of low-voltage-activated t-type calcium channels
    T-type Ca2+ channels were originally called low-voltage-activated (LVA) channels because they can be activated by small depolarizations of the plasma membrane.
  4. [4]
    Structural biology of voltage-gated calcium channels - PMC
    Dec 7, 2023 · The core α1 subunit is self-sufficient for the autonomous function of Cav3 channels, whereas the Cav1 and Cav2 subfamilies necessitate auxiliary ...Introduction · Figure 1 · Structural Architecture Of...
  5. [5]
  6. [6]
  7. [7]
    Molecular Physiology of Low-Voltage-Activated T-type Calcium ...
    There are at least 10 genes encoding α1-subunits of voltage-gated Ca2+ channels (Fig. 1). Alignment of their deduced amino acid sequences suggests that gene ...
  8. [8]
  9. [9]
    Interaction of T-type calcium channel Ca V 3.3 with the β-subunit
    Aug 23, 2010 · ... CaVβ protein played a role in binding with CaV3.3. This is the first demonstration of an α-β subunit interaction in a T-type calcium channel.
  10. [10]
    Interaction of T-type calcium channel Ca(V)3.3 with the β-subunit
    We describe here the identification of a new interaction between Ca(V)3.3 and Ca(V)β proteins. This interaction is of low affinity compared to that between the ...Missing: Cav3 | Show results with:Cav3
  11. [11]
    The α2δ subunits of voltage-gated calcium channels - ScienceDirect
    This review discusses the α 2 δ subunits of voltage-gated calcium channels. The α 2 δ subunits are involved in calcium channel trafficking.
  12. [12]
    Voltage‐gated calcium channels and their auxiliary subunits
    This review will provide a general introduction and then concentrate particularly on the role of auxiliary α2δ subunits in both physiological and pathological ...
  13. [13]
  14. [14]
    Molecular Properties of Voltage-Gated Calcium Channels - NCBI - NIH
    The LVA (or T-type) channels, typically have a small conductance (8-12 pico Siemens (pS)), open in response to small changes from the resting membrane potential ...Cloned Calcium Channels · Low Voltage-Activated (T-Type...
  15. [15]
    A model of the T-type calcium current and the low-threshold spike in ...
    A model of the transient, low-threshold voltage-dependent (T-type) Ca2+ current is constructed using recent whole-cell voltage-clamp data.
  16. [16]
    Zn2+ Sensitivity of High- and Low-Voltage Activated Calcium ...
    The selectivity filter of the pore region of T-type channels has an EEDD locus (29–31), whereas the selectivity filter in high-voltage–gated channels has an ...
  17. [17]
    The Eeee Locus Is the Sole High-Affinity Ca2+ Binding Structure in ...
    Abstract. Selective permeability in voltage-gated Ca2+ channels is dependent upon a quartet of pore-localized glutamate residues (EEEE locus).
  18. [18]
    Ion Trafficking through T-type Ca2+ Channels - PubMed Central
    Voltage-gated Ca2+ channels play a key role in controlling Ca2+ entry during cell depolarization. At least 10 genes encode the main (α1) subunits of ...
  19. [19]
    Ion Interactions in the High-Affinity Binding Locus of a Voltage-Gated ...
    Ion selectivity in Ca2+ channels is based on interactions between permeant ions and the EEEE locus: in a mixture of ions, all of which can pass through the pore ...
  20. [20]
    Properties of membrane currents in isolated smooth muscle cells ...
    ... divalent cations through the Ca2+ channel was Ba2+ greater than Sr2+ approximately Ca2+. Cd2+ blocked the Ca2+ current more effectively than Ni2+. These ...
  21. [21]
    T-type Ca2+ Channels and Pharmacological Blockade - PubMed
    T-type Ca2+ channels are weakly blocked by standard Ca2+ antagonists. Pharmacological blockers are scarce and often lack specificity and/or potency.
  22. [22]
    Gating Kinetics of the α1i T-Type Calcium Channel - PMC
    Channel activation was clearly detectable by −60 mV, and the peak inward current was near −30 mV. The instantaneous I-V relationship (Fig. 1 C, squares) ...
  23. [23]
    Structure, gating, and pharmacology of human Ca V 3.3 channel
    Apr 19, 2022 · Three T-type calcium channel isoforms, including α1G (CaV3.1), α1H (CaV3.2), and α1I (CaV3.3), have been identified in mammals and well ...
  24. [24]
    A cooperative knock-on mechanism underpins Ca2+-selective ... - NIH
    We show that Ca 2+ -selective TRPV channel permeation occurs by a three-binding site knock-on mechanism, whereas a two-binding site knock-on mechanism is ...
  25. [25]
    Phosphorylation of the Cav3.2 T-type calcium channel ... - PNAS
    Oct 19, 2015 · These channels are low voltage-activated calcium channels that play a key role in cellular excitability and various physiological functions.Missing: evolutionary | Show results with:evolutionary
  26. [26]
    Temperature-dependent Modulation of CaV3 T-type Calcium ...
    In this study, we demonstrate that protein kinase A (PKA) and PKC (but not PKG) activation induces a potent increase in Ca V 3.1, Ca V 3.2, and Ca V 3.3 ...
  27. [27]
    Voltage-Gated T-Type Calcium Channel Modulation by Kinases and ...
    Jan 31, 2023 · TTCCs are LVA channels that belong to the family of VGCCs. There are three isoforms of TTCCs, Cav3.1, Cav3.2, and Cav3.3, that are encoded by ...
  28. [28]
    Modulation of Cav3.2 T-type calcium channel permeability by ... - NIH
    Discussion. In the present study, we provide evidence for an essential role of N-glycosylation in the control of human Cav3.2 T-type channel gating.
  29. [29]
    Glycosylation of voltage-gated calcium channels in health and disease
    Surface expression and function of Cav3.2 T-type calcium channels are controlled by asparagine-linked glycosylation. Pflugers Arch. (2013), pp. 1159-1170.
  30. [30]
    Role of the Ubiquitin System in Chronic Pain - Frontiers
    2 T-type calcium channels. This can reduce the ubiquitination of Cav3.2 channels and upregulate Cav3.2 channels, making them more stable on the cell surface.
  31. [31]
    SUMOylation regulates USP5-Cav3.2 calcium channel interactions
    Aug 27, 2019 · Here we describe the regulation of the Cav3.2-USP5 interaction by SUMOylation. We show that endogenous USP5 protein expressed in dorsal root ...
  32. [32]
  33. [33]
  34. [34]
    Calmodulin regulates Cav3 T-type channels at their gating brake
    Dec 8, 2017 · Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif.
  35. [35]
    Dynamics of Low-Threshold Spike Activation in Relay Neurons ... - NIH
    The low-threshold spike (LTS), generated by the transient Ca2+ current IT, plays a pivotal role in thalamic relay cell responsiveness and thus in the nature ...
  36. [36]
    Dendritic Low-Threshold Calcium Currents in Thalamic Relay Cells
    The low-threshold calcium current (IT) underlies burst generation in thalamocortical (TC) relay cells and plays a central role in the genesis of synchronized ...
  37. [37]
    T-Type Calcium Channels Contribute to Burst Firing in a ...
    Aug 11, 2020 · This study characterizes a low-voltage-activated T-type Ca2+ in mediating a unique mode of neuronal firing in medial habenular neurons.
  38. [38]
    T-type Ca2 + channels in absence epilepsy - ScienceDirect.com
    Dysfunction of T-type channels has been strongly implicated in various neurological disorders, including sleep disorders, absence epilepsy, neuropathic pain, ...
  39. [39]
    T-type calcium channels in synaptic plasticity - PMC - NIH
    Here, we review a number of studies showing that T-type channels participate to numerous homo- and hetero-synaptic plasticity mechanisms.
  40. [40]
    T-Type Ca2+ Channels Mediate a Critical Period of Plasticity in ...
    Apr 10, 2024 · Adult-born granule cells (abGCs) exhibit a transient period of elevated synaptic plasticity that plays an important role in hippocampal function ...Electrophysiology · Gabaergic Inhibition... · T-Type Ca Channels Are Allow...
  41. [41]
    Central and peripheral contributions of T-type calcium channels in ...
    May 2, 2022 · Recent evidence suggests that T-type channels contribute to excitability of neurons all along the ascending and descending pain pathways.Missing: auxiliary 2023<|control11|><|separator|>
  42. [42]
    The T-type Ca2+ Channel Cav3.2 Regulates Differentiation of ...
    Mar 15, 2019 · Gene transcription and splicing of T-type channels are evolutionarily-conserved strategies for regulating channel expression and gating. Plos ...
  43. [43]
    Mechanisms of neuromodulatory volume transmission - Nature
    May 24, 2024 · In contrast, the CaV2-dependence of dopamine release is partial, and CaV1 (L-type) and CaV3 (T-type) channels also contribute [32, 71]. Dopamine ...
  44. [44]
    Functional role of voltage gated Ca2+ channels in heart automaticity
    Feb 2, 2015 · In the “calcium clock” model of pacemaking the key mechanism in the diastolic depolarization is a spontaneous rhythmic phenomenon of Ca2+ ...
  45. [45]
  46. [46]
    [PDF] THE POSSIBLE PROARRHYTHMIC EFFECTS OF SOME NON ...
    cardiac Purkinje fibers and papillary muscles. ... T–type calcium channels (ICaT/Cav3.1–3.3) (Ross ... increased dispersion of repolarization and triangulation of ...
  47. [47]
    Functional role of voltage gated Ca2+ channels in heart automaticity
    Genomic analysis demonstrated that Cav1.3 and Cav3.1 channels are widely expressed in pacemaker tissue of mice, rabbits and humans. Importantly, human ...
  48. [48]
    Species Difference in the Contribution of T-Type Calcium Current to ...
    The contribution of the T-type Ca2+ current to cardiac pacemaking was examined in isolated right atrial tissue from the mouse, guinea pig, and rabbit using ...Missing: rodent | Show results with:rodent
  49. [49]
    T-Type Calcium Current Contributes to Escape Automaticity and ...
    Jul 24, 2013 · We conclude that T-type Ca2+ channels play an important role in ventricular escape automaticity and protect the heart against excess bradycardia ...
  50. [50]
    The Dysfunction of Ca2+ Channels in Hereditary and Chronic ...
    Oct 27, 2023 · In this review, I discuss the origin of genetic Ca 2+ channelopathies of L- and T-type voltage-gated calcium channels in humans and the role of the non-genetic ...
  51. [51]
    Increasing T‐type calcium channel activity by β‐adrenergic ... - NIH
    Cav3.1 T‐type Ca2+ channel current (ICa‐T) contributes to heart rate genesis but is not known to contribute to heart rate regulation by the ...Missing: rodent | Show results with:rodent
  52. [52]
    Calcium Channels in Postnatal Development of Rat Pancreatic Beta ...
    Others have also shown that the inhibition of T-type calcium channels reduced insulin secretion by 60–70% at 6 mM glucose in human pancreatic beta cells (46).
  53. [53]
    The T-type calcium channel CaV3.2 regulates insulin secretion in ...
    Oct 31, 2022 · Voltage-gated Ca 2+ (Ca V ) channel dysfunction leads to impaired glucose-stimulated insulin secretion in pancreatic β-cells and contributes ...
  54. [54]
    Voltage-Gated Ion Channels in Human Pancreatic β-Cells
    Jun 1, 2008 · Voltage-gated T-type and L-type Ca 2+ channels as well as Na + channels participate in glucose-stimulated electrical activity and insulin secretion.
  55. [55]
    T-type Ca2+ channels in vascular smooth muscle: Multiple functions
    T-type channels may generate pacemaker activity, are linked to cell proliferation, and may have tissue-specific functions in vascular smooth muscle.
  56. [56]
  57. [57]
    T-Type Voltage-Gated Calcium Channels: Potential Regulators of ...
    This review provides a detailed examination of T-type voltage-gated calcium channels, highlighting their structure, electrophysiology, biophysics, expression ...Missing: seminal | Show results with:seminal<|control11|><|separator|>
  58. [58]
    Activation of mouse sperm T-type Ca2+ channels by adhesion to the ...
    Abstract. The sperm acrosome reaction is a Ca2+-dependent exocytotic event that is triggered by adhesion to the mammalian egg's zona pellucida.
  59. [59]
    T-type Ca2+ channels in sperm function - PubMed - NIH
    Amongst these channels, the voltage dependent Ca2+ channels (CaV) of the T-type (CaV3) appear to have an eminent role in the acrosome reaction (AR) of some ...Missing: 2025 study
  60. [60]
    Blocking T-type calcium channels disrupts spermatogenesis in vivo ...
    Blocking T-type calcium channels have been demonstrated to disrupt normal spermatogenesis and steroidogenesis, potentially resulting in male infertility [1].
  61. [61]
    Calcium and bone disease - PMC - PubMed Central - NIH
    Both bone forming and bone resorbing cells use calcium signals as regulators of differentiation and activity. This has been studied in more detail in ...
  62. [62]
    [PDF] Examining Mechanisms for Voltage-Sensitive Calcium Channel
    Jul 1, 2023 · Blocking expression of the T-type channel Cav3.2 using short hairpin RNAs in mouse bone marrow-derived macrophages significantly suppressed.
  63. [63]
    Large conductance voltage-and calcium-activated K+ (BK) channel ...
    This model considers multiple ionic conductances, including calcium channels (L-type and T-type), voltage-gated potassium channels (Kv1, KCNQ), ATP ...
  64. [64]
  65. [65]
    Ca2+ influx via T-type channels modulates PDGF-induced ...
    Overexpression of T‐type calcium channels in HEK‐293 cells increases intracellular calcium without affecting cellular proliferation. 24 July 2000 | FEBS ...
  66. [66]
    T-type calcium channels in differentiation and proliferation
    Low-voltage activated, T-type calcium channels (T-channels) are expressed in many developing tissues and may be important in regulating important cellular ...
  67. [67]
    Structure and alternative splicing of the gene encoding α 1G , a ...
    This report describes the structure of CACNA1G and presents evidence for alternative splicing involving six exons.
  68. [68]
    Expanded alternative splice isoform profiling of the mouse Cav3.1 ...
    May 29, 2009 · ... C-terminal tail of the channel. Splicing at the C-terminus of Cav3.3/α1I T-type channels also alters electrophysiological properties, as the ...
  69. [69]
    o43497 · cac1g_human - UniProt
    Highly expressed in brain, in particular in the amygdala, subthalamic nuclei, cerebellum and thalamus. Moderate expression in heart; low expression in placenta, ...
  70. [70]
    T-Type Calcium Channels: A Mixed Blessing - MDPI
    This review puts into context the relevance of T-type calcium channels in cancer and in chemotherapy side effects, considering also the cardiotoxicity.Missing: auxiliary channelosome
  71. [71]
    Molecular Physiology of Low-Voltage-Activated T-type Calcium ...
    Electrophysiological studies of recombinant chan- nels show that Cav3.1 (formerly α1G) and Cav3.2 (α1H) have similar activation and inactivation kinetics, but ...
  72. [72]
    and Cav3 subtype-dependent alterations in T-type calcium channel ...
    The major limiting factor to a contribution by Cav3.2 and Cav3.3 to rebound firing at 21 °C was a slow recovery from inactivation, which prevented the current ...
  73. [73]
    Thalamic T-type Ca2+ channels and NREM sleep - PubMed Central
    T-type Ca 2+ channels constitute the single, most crucial, voltage-dependent conductance that permeates all major NREM sleep oscillations in TC and NRT ...
  74. [74]
    Altered thalamocortical rhythmicity and connectivity in mice ... - PNAS
    Jun 8, 2015 · Our findings indicate that Ca V 3.1 KO mice displayed attenuated low-frequency oscillations in thalamocortical loops, especially in the 1- to 4-Hz delta band.
  75. [75]
    Genetic T-type calcium channelopathies
    In this short review, we present the genetics of T-type channels with an emphasis on structure-function relationships and associated channelopathies.
  76. [76]
    Functional Characterization and Neuronal Modeling of the Effects of ...
    May 11, 2005 · Eleven SNPs altered some aspect of channel gating. Computer simulations predict that seven of the SNPs would increase firing of neurons, with ...
  77. [77]
    Reticular thalamic hyperexcitability drives autism spectrum disorder ...
    Aug 20, 2025 · Our findings provide the evidence of RT hyperexcitability contributing to ASD-related behaviors and highlight the RT as a promising therapeutic ...
  78. [78]
    Cacna1g - SFARI Gene
    Human Gene Module / Chromosome 17 / CACNA1G. CACNA1Gcalcium channel, voltage ... chromosomal region linked to autism identifies CACNA1G as a novel candidate gene ...
  79. [79]
    607904 - CALCIUM CHANNEL, VOLTAGE-DEPENDENT, T TYPE ...
    (1998) mapped the CACNA1H gene to chromosome 16p13.3. By interspecific backcross analysis, they mapped the mouse Cacna1h gene to chromosome 17. ▻ Gene Function.
  80. [80]
    A profile of alternative RNA splicing and transcript variation of ...
    T-type, low-voltage-activated calcium channels appear to play significant and possibly obligatory roles in idiopathic generalized epilepsies (IGEs) (5,6).
  81. [81]
    Expression and Regulation of Cav3.2 T-Type Calcium Channels ...
    We describe a novel finding that Cav3. 2 T-type calcium channels are involved in the development and maintenance of the inflammatory hyperalgesia induced by ...
  82. [82]
    Genetic Tracing of Cav3.2 T-Type Calcium Channel Expression in ...
    As development progresses the number of DRG cells expressing the Cav3.2 channel reaches around 7% of the DRG at E16.5, and remains constant until E18.5.
  83. [83]
    [PDF] Targeting T-type/CaV3.2 channels for chronic pain
    Likewise,. Cav3.1 and CaV3.2 have fast kinetic properties (unlike Cav3.3), their activation and inactivation are both rapid compared to HVA calcium channels ...<|separator|>
  84. [84]
    T-type calcium channel modulation by hydrogen sulfide in ... - Frontiers
    The present review provides relevant information about H 2 S modulation on the Ca v 3.2 T-type channels in neuropathic pain conditions.
  85. [85]
    CACNA1H mutations in autism spectrum disorders - PubMed - NIH
    Here, we identify missense mutations in the calcium channel gene CACNA1H (T-type Ca(V)3.2) in 6 of 461 individuals with ASD.
  86. [86]
    Contribution of CACNA1H Variants in Autism Spectrum Disorder ...
    Mar 7, 2022 · CACNA1H is reported to be a strong candidate for ASD as both de novo and inherited rare variants in CACNA1H were identified in individuals with ...
  87. [87]
    Icariside II, a Prenyl-Flavonol, Alleviates Inflammatory and ...
    Apr 28, 2023 · Cav3.2 channels are upregulated during neuropathic pain or peripheral inflammation in part due to an increased association with the ...
  88. [88]
  89. [89]
    The CaV3.3 calcium channel is the major sleep spindle pacemaker ...
    Aug 16, 2011 · CaV3.3 protein is expressed abundantly in the nucleus reticularis thalami (nRt), an essential oscillatory burst generator. We report the ...Missing: pituitary respiratory
  90. [90]
    CaV 3.1 and CaV 3.3 account for T-type Ca2+ current in GH3 cells
    We report here the electrophysiological and molecular characteristics of the whole-cell Ca2+ current in GH3 clonal pituitary cells. The current inactivation at ...
  91. [91]
    Interdependence of cellular and network properties in respiratory ...
    May 1, 2024 · We propose a unifying, data-driven model of respiratory rhythm generation, which bridges the gap between these competing theories.Missing: Cav3. | Show results with:Cav3.
  92. [92]
    Ca v 3 T-type calcium channels - Wiley Online Library
    Feb 28, 2012 · T-type channels are unique among the voltage-gated calcium channels in their fast kinetics and low voltages of activation and inactivation, ...
  93. [93]
    The potential roles of T-type Ca2+ channels in motor ... - Frontiers
    Oct 27, 2013 · T-type Ca2+ channels mediate intrinsic neuronal oscillations and rhythmic burst spiking, and facilitate the generation of tremor rhythms in ...
  94. [94]
    CACNA1I gain-of-function mutations differentially affect channel ...
    Mar 11, 2021 · Here, we examined four heterozygous missense variants in CACNA1I, encoding the Ca v 3.3 channel, in patients with variable neurodevelopmental phenotypes.
  95. [95]
    Uncovering convergence and divergence between autism ... - Nature
    Sep 5, 2024 · Schizophrenia, autism spectrum disorders and developmental disorders share specific disruptive coding mutations. Nat Commun. 2021;12:5353 ...<|separator|>
  96. [96]
    Concomitant genetic ablation of L-type Cav1.3 (α1D) and T ... - Nature
    Nov 3, 2020 · We show that concomitant loss of SAN-VGCCs prevents physiological SAN automaticity, blocks impulse conduction and compromises ventricular rhythmicity.
  97. [97]
    T-type calcium channels and vascular function: the new kid on ... - NIH
    ... T-type channel pore forming α1 subunit with α2δ and γ subunits. However, these auxiliary subunits still appear capable of modulating T-type activity. Thus ...
  98. [98]
    Ion channel molecular complexes in vascular smooth muscle
    Multiple ion channels regulate VSM contraction by controlling membrane potential and the magnitude of intracellular calcium concentration [Ca2+]i (Knot and ...
  99. [99]
    L- and T-type calcium channels control aldosterone production from ...
    We conclude that T- and L-type calcium channels play equally important roles in controlling aldosterone production from human adrenals.
  100. [100]
    T-Type Calcium Channel: A Privileged Gate for Calcium Entry and ...
    T-type calcium channels are particularly well suited for responding under these conditions and conveying calcium into the cell, at the right place for ...
  101. [101]
    Aldosterone Increases T-Type Calcium Currents in Human ...
    In human adrenocarcinoma cells, aldosterone increases, through an autocrine pathway, the expression of T-type calcium channels and therefore modifies the ...
  102. [102]
    N-/T-Type vs. L-Type Calcium Channel Blocker in Treating Chronic ...
    Feb 22, 2023 · The inhibition of T-type calcium channels dilates afferent and efferent arterioles, as well as alleviates glomerular damage [41]. It has been ...
  103. [103]
    Targeting ion channel networks in diabetic kidney disease
    Jun 26, 2025 · This review systematically examines the roles of ion channels in glomerular filtration barrier dysfunction, tubular reabsorption, and fibrotic processes in DKD
  104. [104]
    N-/T-Type vs. L-Type Calcium Channel Blocker in Treating Chronic ...
    This meta-analysis of 12 RCTs with 967 CKD patients who were treated with RAS inhibitors demonstrated that, when compared with L-type CCB, N-/T- ...
  105. [105]
    Role of Voltage-Gated Calcium Channels in Pancreatic β-Cell ...
    Oct 1, 2006 · T-type calcium channels facilitate insulin secretion by enhancing general excitability in the insulin-secreting β-cell line, INS-1.Introduction · III. Role of CaV Channels in β... · IV. Role of CaV Channels in β...<|separator|>
  106. [106]
    Ca 2+ signaling and metabolic stress-induced pancreatic β-cell failure
    Early in the development of Type 2 diabetes (T2D), metabolic stress brought on by insulin resistance and nutrient overload causes β-cell hyperstimulation.Abstract · Metabolic stimuli-insulin... · Effects of metabolic stress... · Discussion
  107. [107]
    Nickel block of three cloned T-type calcium channels - PubMed - NIH
    Nickel has been proposed to be a selective blocker of low-voltage-activated, T-type calcium channels. ... low micromolar concentrations (IC(50) = 13 microM).
  108. [108]
    T-type calcium channel antagonists, mibefradil and NNC-55-0396 ...
    May 21, 2015 · This regulation of Ca2+ homeostasis allows T-type Ca2+ channels to control cell proliferation and apoptosis, or death. There are increasing data ...
  109. [109]
    Withdrawal of Posicor From Market | Circulation
    The withdrawal came after reports of dangerous and even fatal interactions with at least 25 other drugs, including common antibiotics, antihistamines, and ...Missing: reason | Show results with:reason
  110. [110]
    Recent advances in the development of T‐type calcium channel ...
    Cav3.2 T‐type calcium channels are important regulators of pain signals in the afferent pain pathway, and their activities are dysregulated during various ...
  111. [111]
    Contributions of T-Type Voltage-Gated Calcium Channels to ...
    T-type channels have now been shown to underlie regenerative low-threshold spikes and burst firing in neurons throughout the central nervous system (CNS), ...
  112. [112]
    Subunit-specific modulation of T-type calcium channels by zinc - PMC
    The modulatory effects on T-channels occurs at Zn2+ concentrations that are of physiological relevance in the CNS and are likely to have an impact on neuron ...Missing: agonist levels
  113. [113]
    T-type calcium channel modulation by hydrogen sulfide in ...
    This review provides relevant information about hydrogen sulfide modulation on Cav3.2, the main T-type calcium channel subunit participating in neuropathic pain ...
  114. [114]
    Calcium channels in anesthesia management: A molecular and ...
    May 10, 2025 · Anesthetics, particularly propofol, modulate calcium influx through ligand-gated calcium channels, impacting both central nervous system and ...<|control11|><|separator|>
  115. [115]
    Recent advances in the development of T‐type calcium channel ...
    Jun 13, 2017 · Here, we review recent developments in the discovery of novel classes of T-type calcium channel blockers, and their analgesic effects in animal models of pain ...
  116. [116]
    Painful diabetic neuropathy: The role of ion channels - ScienceDirect
    Cav channels. The N-type (Cav2.2) and T-type (Cav3.2) are the main channels targeted for the development of calcium channel antagonists. N-type calcium ...
  117. [117]
    Ethosuximide - StatPearls - NCBI Bookshelf - NIH
    Ethosuximide is FDA-approved for the management of absence seizures in patients 3 or older. · Ethosuximide has demonstrated level A evidence for managing ...
  118. [118]
    The Role of T-Type Calcium Channel Genes in Absence Seizures
    Here, we review the role of T-type calcium channel genes in the pathogenesis of absence seizures, and emphasize that variants of these genes may initiate ...
  119. [119]
    Cinnarizine: Uses, Interactions, Mechanism of Action - DrugBank
    Jun 13, 2005 · Cinnarizine is a drug used for the management of labyrinthine disorder symptoms, including vertigo, tinnitus, nystagmus, nausea, and vomiting.Identification · Pharmacology · Interactions · Categories
  120. [120]
    Cinnarizine: A Contemporary Review - PMC - PubMed Central
    Apr 25, 2017 · Cinnarizine is approved for nausea, vomiting, motion sickness, inner ear disorders and is considered as first-line pharmacotherapy for management of vertigo.
  121. [121]
    KTt-45, a T-type calcium channel blocker, acts as an anticancer ...
    Dec 13, 2023 · KTt-45 could inhibit cell growth and trigger mitochondrial-dependent apoptosis in HeLa cervical cancer cells.Missing: preclinical | Show results with:preclinical
  122. [122]
    Modulation of subthalamic T-type Ca 2+ channels remedies ... - JCI
    We therefore conclude that modulation of subthalamic T-type Ca2+ currents and consequent burst discharges may provide new strategies for the treatment of PD.
  123. [123]
    The T-type calcium channel as a new therapeutic target for ...
    Aug 6, 2025 · Local application of T-type Ca(2+) channel blockers into STN would also dramatically decrease the burst discharges and improve parkinsonian ...
  124. [124]
    Targeting T-type channels in cancer: What is on and what is off?
    On-target vs off-target effects of T-type calcium channel (TTCC) blockers used in cancer cells. Selective inhibition of TTCCs (indicated by white cross on inner ...Missing: challenges | Show results with:challenges
  125. [125]
    Rethinking GABAAR-targeting anesthetics | Cell Biology and ...
    Jun 14, 2025 · The neuroprotective properties of these compounds may be partially attributed to their ability to inhibit T-type calcium channels, particularly ...