首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Low voltage-activated T-type calcium (Ca) channels contribute to the normal development of the heart and are also implicated in pathophysiological states such as cardiac hypertrophy. Functionally distinct T-type Ca channel isoforms can be generated by alternative splicing from each of three different T-type genes (CaV3.1, CaV3.2,CaV3 .3), although it remains to be described whether specific splice variants are associated with developmental states and pathological conditions. We aimed to identify and functionally characterize CaV3.2 T-type Ca channel alternatively spliced variants from newborn animals and to compare with adult normotensive and spontaneously hypertensive rats (SHR). DNA sequence analysis of full-length CaV3.2 cDNA generated from newborn heart tissue identified ten major regions of alternative splicing, the more common variants of which were analyzed by quantitative real-time PCR (qRT-PCR) and also subject to functional examination by whole-cell patch clamp. The main findings are that: (1) cardiac CaV3.2 T-type Ca channels are subject to considerable alternative splicing, (2) there is preferential expression ofCaV3 .2(-25) splice variant channels in newborn rat heart with a developmental shift in adult heart that results in approximately equal levels of expression of both (+25) and (-25) exon variants, (3) in the adult stage of hypertensive rats there is a both an increase in overallCaV3 .2 expression and a shift towards expression of CaV3.2(+25) containing channels as the predominant form, and (4) alternative splicing confers a variant-specific voltage-dependent facilitation ofCaV3 .2 channels. We conclude that CaV3.2 alternative splicing generates significant T-type Ca channel structural and functional diversity with potential implications relevant to cardiac developmental and pathophysiological states.  相似文献   

2.
Low voltage-activated T-type calcium (Ca) channels contribute to the normal development of the heart and are also implicated in pathophysiological states such as cardiac hypertrophy. Functionally distinct T-type Ca channel isoforms can be generated by alternative splicing from each of three different T-type genes (CaV3.1, CaV3.2, CaV3.3), although it remains to be described whether specific splice variants are associated with developmental states and pathological conditions. We aimed to identify and functionally characterize CaV3.2 T-type Ca channel alternatively spliced variants from newborn animals and to compare with adult normotensive and spontaneously hypertensive rats (SHR). DNA sequence analysis of full-length CaV3.2 cDNA generated from newborn heart tissue identified ten major regions of alternative splicing, the more common variants of which were analyzed by quantitative real-time PCR (qRT-PCR) and also subject to functional examination by whole-cell patch clamp. The main findings are that: (1) cardiac CaV3.2 T-type Ca channels are subject to considerable alternative splicing, (2) there is preferential expression of CaV3.2(−25) splice variant channels in newborn rat heart with a developmental shift in adult heart that results in approximately equal levels of expression of both (+25) and (−25) exon variants, (3) in the adult stage of hypertensive rats there is both an increase in overall CaV3.2 expression and a shift towards expression of CaV3.2(+25) containing channels as the predominant form and (4) alternative splicing confers a variant-specific voltage-dependent facilitation of CaV3.2 channels. We conclude that CaV3.2 alternative splicing generates significant T-type Ca channel structural and functional diversity with potential implications relevant to cardiac developmental and pathophysiological states.Key words: voltage-dependent facilitation, alternative splicing, T-type calcium channel, hypertension, cardiac hypertrophy  相似文献   

3.
The regulation of intracellular Ca2+ is essential for cardiomyocyte function, and alterations in proteins that regulate Ca2+ influx have dire consequences in the diseased heart. Low voltage-activated, T-type Ca2+ channels are one pathway of Ca2+ entry that is regulated according to developmental stage and in pathological conditions in the adult heart. Cardiac T-type channels consist of two main types, Cav3.1 (α1G) and Cav3.2 (α1H), and both can be induced in the myocardium in disease and injury but still, relatively little is known about mechanisms for their regulation and their respective functions. This article integrates previous data establishing regulation of T-type Ca2+ channels in animal models of cardiac disease, with recent data that begin to address the functional consequences of cardiac Cav3.1 and Cav3.2 Ca2+ channel expression in the pathological setting. The putative association of T-type Ca2+ channels with Ca2+ dependent signaling pathways in the context of cardiac hypertrophy is also discussed.  相似文献   

4.
L-type voltage gated Ca2+ channels are considered to be the primary source of calcium influx during the myogenic response. However, many vascular beds also express T-type voltage gated Ca2+ channels. Recent studies suggest that these channels may also play a role in autoregulation. At low pressures (40–80 mmHg) T-type channels affect myogenic responses in cerebral and mesenteric vascular beds. T-type channels also seem to be involved in skeletal muscle autoregulation. This review discusses the expression and role of T-type voltage gated Ca2+ channels in the autoregulation of several different vascular beds. Lack of specific pharmacological inhibitors has been a huge challenge in the field. Now the research has been strengthened by genetically modified models such as mice lacking expression of T-type voltage gated Ca2+ channels (CaV3.1 and CaV3.2). Hopefully, these new tools will help further elucidate the role of voltage gated T-type Ca2+ channels in autoregulation and vascular function.  相似文献   

5.
The regulation of arterial tone is critical in the spatial and temporal control of cerebral blood flow. Voltage-gated Ca2+ (CaV) channels are key regulators of excitation–contraction coupling in arterial smooth muscle, and thereby of arterial tone. Although L- and T-type CaV channels have been identified in rodent smooth muscle, little is known about the expression and function of specific CaV subtypes in human arteries. Here, we determined which CaV subtypes are present in human cerebral arteries and defined their roles in determining arterial tone. Quantitative polymerase chain reaction and Western blot analysis, respectively, identified mRNA and protein for L- and T-type channels in smooth muscle of cerebral arteries harvested from patients undergoing resection surgery. Analogous to rodents, CaV1.2 (L-type) and CaV3.2 (T-type) α1 subunits were expressed in human cerebral arterial smooth muscle; intriguingly, the CaV3.1 (T-type) subtype present in rodents was replaced with a different T-type isoform, CaV3.3, in humans. Using established pharmacological and electrophysiological tools, we separated and characterized the unique profiles of Ca2+ channel subtypes. Pressurized vessel myography identified a key role for CaV1.2 and CaV3.3 channels in mediating cerebral arterial constriction, with the former and latter predominating at higher and lower intraluminal pressures, respectively. In contrast, CaV3.2 antagonized arterial tone through downstream regulation of the large-conductance Ca2+-activated K+ channel. Computational analysis indicated that each Ca2+ channel subtype will uniquely contribute to the dynamic regulation of cerebral blood flow. In conclusion, this study documents the expression of three distinct Ca2+ channel subtypes in human cerebral arteries and further shows how they act together to orchestrate arterial tone.  相似文献   

6.
T-type Ca2+ channel family includes three subunits CaV3.1, CaV3.2 and CaV3.3 and have been shown to control burst firing and intracellular Ca2+ concentration ([Ca2+]i) in neurons. Here, we investigated whether CaV3.1 channels could generate a pacemaker current and contribute to cell excitability. CaV3.1 clones were over-expressed in the neuronal cell line NG108-15. CaV3.1 channel expression induced repetitive action potentials, generating spontaneous membrane potential oscillations (MPOs) and concomitant [Ca2+]i oscillations. These oscillations were inhibited by T-type channels antagonists and were present only if the membrane potential was around −61 mV. [Ca2+]i oscillations were critically dependent on Ca2+ influx through CaV3.1 channels and did not involve Ca2+ release from the endoplasmic reticulum. The waveform and frequency of the MPOs are constrained by electrophysiological properties of the CaV3.1 channels. The trigger of the oscillations was the CaV3.1 window current. This current induced continuous [Ca2+]i increase at −60 mV that depolarized the cells and triggered MPOs. Shifting the CaV3.1 window current potential range by increasing the external Ca2+ concentration resulted in a corresponding shift of the MPOs threshold. The hyperpolarization-activated cation current (Ih) was not required to induce MPOs, but when expressed together with CaV3.1 channels, it broadened the membrane potential range over which MPOs were observed. Overall, the data demonstrate that the CaV3.1 window current is critical in triggering intrinsic electrical and [Ca2+]i oscillations.  相似文献   

7.
We investigated the biophysical mechanism of inhibition of recombinant T-type calcium channels CaV3.1 and CaV3.2 by nitrous oxide (N2O). To identify functionally important channel structures, chimeras with reciprocal exchange of the N-terminal domains I and II and C-terminal domains III and IV were examined. In whole-cell recordings N2O significantly inhibited CaV3.2, and – less pronounced – CaV3.1. A CaV3.2-prevalent inhibition of peak currents was also detected in cell-attached multi-channel patches. In cell-attached patches containing ≤3 channels N2O reduced average peak current of CaV3.2 by decreasing open probability and open time duration. Effects on CaV3.1 were smaller and mediated by a reduced fraction of sweeps containing channel activity. Without drug, single CaV3.1 channels were significantly less active than CaV3.2. Chimeras revealed that domains III and IV control basal gating properties. Domains I and II, in particular a histidine residue within CaV3.2 (H191), are responsible for the subtype-prevalent N2O inhibition. Our study demonstrates the biophysical (open times, open probability) and structural (domains I and II) basis of action of N2O on CaV3.2. Such a fingerprint of single channels can help identifying the molecular nature of native channels. This is exemplified by a characterization of single channels expressed in human hMTC cells as functional homologues of recombinant CaV3.1.  相似文献   

8.
9.
Voltage-gated calcium channels (VGCCs) regulate calcium influx into all excitable cells. In the heart, the main calcium channels are the L-type VGCCs (LTCCs). These are localised to the sarcolemmal membrane, and are hetero-oligomeric complexes comprised of three non-covalently associated polypeptides; alpha1 (CaV1.2), alpha2delta and beta. We recently reported the 3D structure for a monomeric form of the cardiac LTCC1 using electron microscopy and single particle analysis. We also determined the first medium/low resolution structure of a T-type voltage gated calcium channel (CaV3.1) polypeptide. We identified the transmembrane and cytoplasmic domains of the T-type channel using labelling studies to determine the position of the C-terminus. By modelling of the CaV3.1 structure (comparable at these resolutions to CaV1.2) into the cardiac LTCC volume, we were able to delineate the subunit boundaries of the cardiac LTCC, leading to a proposal for a putative orientation of the LTCC with respect to the membrane bilayer. We have now extended these studies to include labelling of the extracellular alpha2 polypeptide using affinity purified antibodies raised against the Von Willebrand Factor A (VWA) domain and calmodulin-gold labelling of the C-terminus of CaV1.2. These data provide further support for the proposed orientation of the 3D structure of the cardiac LTCC.  相似文献   

10.
11.
We used MCF-7 human breast cancer cells that endogenously express Cav3.1 and Cav3.2 T-type Ca2+ channels toward a mechanistic study on the effect of EGCG on [Ca2+]i. Confocal Ca2+ imaging showed that EGCG induces a [Ca2+]i spike which is due to extracellular Ca2+ entry and is sensitive to catalase and to low-specificity (mibefradil) and high-specificity (Z944) T-type Ca2+channel blockers. siRNA knockdown of T-type Ca2+ channels indicated the involvement of Cav3.2 but not Cav3.1. Application of EGCG to HEK cells expressing either Cav3.2 or Cav3.1 induced enhancement of Cav3.2 and inhibition of Cav3.1 channel activity. Measurements of K+ currents in MCF-7 cells showed a reversible, catalase-sensitive inhibitory effect of EGCG, while siRNA for the Kv1.1 K+ channel induced a reduction of the EGCG [Ca2+]i spike. siRNA for Cav3.2 reduced EGCG cytotoxicity to MCF-7 cells, as measured by calcein viability assay. Together, data suggest that EGCG promotes the activation of Cav3.2 channels through K+ current inhibition leading to membrane depolarization, and in addition increases Cav3.2 currents. Cav3.2 channels are in part responsible for EGCG inhibition of MCF-7 viability, suggesting that deregulation of [Ca2+]i by EGCG may be relevant in breast cancer treatment.  相似文献   

12.
Voltage-gated Ca2+ channels (VGCCs) are recognized for their superb ability for the preferred passage of Ca2+ over any other more abundant cation present in the physiological saline. Most of our knowledge about the mechanisms of selective Ca2+ permeation through VGCCs was derived from the studies on native and recombinant L-type representatives. However, the specifics of the selectivity and permeation of known recombinant T-type Ca2+-channel α1 subunits, Cav3.1, Cav3.2 and Cav3.3, are still poorly defined. In the present study we provide comparative analysis of the selectivity and permeation Cav3.1, Cav3.2, and Cav3.3 functionally expressed in Xenopus oocytes. Our data show that all Cav3 channels select Ca2+ over Na+ by affinity. Cav3.1 and Cav3.2 discriminate Ca2+, Sr2+ and Ba2+ based on the ion's effects on the open channel probability, whilst Cav3.3 discriminates based on the ion's intrapore binding affinity. All Cav3s were characterized by much smaller difference in the KD values for Na+ current blockade by Ca2+ (KD1 ∼ 6 μM) and for Ca2+ current saturation (KD2 ∼ 2 mM) as compared to L-type channels. This enabled them to carry notable mixed Na+/Ca2+ current at close to physiological Ca2+ concentrations, which was the strongest for Cav3.3, smaller for Cav3.2 and the smallest for Cav3.1. In addition to intrapore Ca2+ binding site(s) Cav3.2, but not Cav3.1 and Cav3.3, is likely to possess an extracellular Ca2+ binding site that controls channel permeation. Our results provide novel functional tests for identifying subunits responsible for T-type Ca2+ current in native cells.  相似文献   

13.
Low voltage-activated (LVA) T-type calcium channels play critical roles in the excitability of many cell types and are a focus of research aimed both at understanding the physiological basis of calcium channel-dependent signaling and the underlying pathophysiology associated with hyperexcitability disorders such as epilepsy. These channels play a critical role towards neuronal firing in both conducting calcium ions during action potentials and also in switching neurons between distinct modes of firing. In this review the properties of the CaV3.1, CaV3.2 and CaV3.3 T-type channel isoforms is discussed in relation to their individual contributions to action potentials during burst and tonic firing states as well their roles in switching between firing states.  相似文献   

14.
Voltage-gated Ca2+ (CaV) channels are transmembrane proteins comprising three subfamilies named CaV1, CaV2 and CaV3. The CaV3 channel subfamily groups the low-voltage activated Ca2+ channels (LVA or T-type) a significant role in regulating neuronal excitability. CaV3 channel activity may lead to the generation of complex patterns of action potential firing such as the postinhibitory rebound (PIR). In the adult spinal cord, these channels have been found in dorsal horn interneurons where they control physiological events near the resting potential and participate in determining excitability. In motoneurons, CaV3 channels have been found during development, but their functional expression has not yet been reported in adult animals. Here, we show evidence for the presence of CaV3 channel-mediated PIR in motoneurons of the adult turtle spinal cord. Our results indicate that Ni2+ and NNC55-0396, two antagonists of CaV3 channel activity, inhibited PIR in the adult turtle spinal cord. Molecular biology and biochemical assays revealed the expression of the CaV3.1 channel isotype and its localization in motoneurons. Together, these results provide evidence for the expression of CaV3.1 channels in the spinal cord of adult animals and show also that these channels may contribute to determine the excitability of motoneurons.  相似文献   

15.
Both N- and T-type calcium ion channels have been implicated in pain transmission and the N-type channel is a well-validated target for the treatment of neuropathic pain. An SAR investigation of a series of substituted aminobenzothiazoles identified a subset of five compounds with comparable activity to the positive control Z160 in a FLIPR-based intracellular calcium response assay measuring potency at both CaV2.2 and CaV3.2 channels. These compounds may form the basis for the development of drug leads and tool compounds for assessing in vivo effects of variable modulation of CaV2.2 and CaV3.2 channels.  相似文献   

16.
Calcium entry through voltage-gated calcium channels has widespread cellular effects upon a host of physiological processes including neuronal excitability, muscle excitation-contraction coupling, and secretion. Using single particle analysis methods, we have determined the first three-dimensional structure, at 23 Å resolution, for a member of the low voltage-activated voltage-gated calcium channel family, CaV3.1, a T-type channel. CaV3.1 has dimensions of ∼115 × 85 × 95 Å, composed of two distinct segments. The cytoplasmic densities form a vestibule below the transmembrane domain with the C terminus, unambiguously identified by the presence of a His tag being ∼65 Å long and curling around the base of the structure. The cytoplasmic assembly has a large exposed surface area that may serve as a signaling hub with the C terminus acting as a “fishing rod” to bind regulatory proteins. We have also determined a three-dimensional structure, at a resolution of 25 Å, for the monomeric form of the cardiac L-type voltage-gated calcium (high voltage-activated) channel with accessory proteins β and α2δ bound to the ion channel polypeptide CaV1.2. Comparison with the skeletal muscle isoform finds a good match particularly with respect to the conformation, size, and shape of the domain identified as that formed by α2. Furthermore, modeling of the CaV3.1 structure (analogous to CaV1.2 at these resolutions) into the heteromeric L-type voltage-gated calcium channel complex volume reveals multiple interaction sites for β-CaV1.2 binding and for the first time identifies the size and organization of the α2δ polypeptides.To date, five different types of voltage-gated calcium channels (VGCCs)4 have been identified, L, N, P/Q, R, and T, and classified according to their physiological and pharmacological characteristics (13). On the basis of their electrophysiological properties, VGCCs can be divided into two classes: high voltage-activated (HVA) and low voltage-activated (LVA). T-type Ca2+ channels form the LVA family and are characterized by their low threshold of activation, small single channel conductance, slow deactivation, and a low sensitivity to classical blockers of HVA channels (46). T-type channels have a central role regulating, for example, cardiac pacemaking of sinoatrial node cells and tonic firing patterns in neurons (5). Three T-type channel isoforms have been identified and cloned: CaV3.1, CaV3.2, and CaV3.3, with each isoform possessing several splice variants showing distinct functional properties (reviewed in Ref. 7).Each VGCC is composed of a pore-forming polypeptide termed the CaV α1-subunit, with 10 mammalian α1 isoforms identified, divided into three subfamilies: CaV1–3 (8). Housed within the CaV α1 subunit are the calcium pore, voltage-sensing apparatus, drug binding sites, and numerous structural determinants required for binding auxiliary subunits and other regulatory proteins. Analysis of the amino acid sequences and predicted secondary structure of the T-type channels suggests a similar topology to the HVA CaV α1 subunits and K+ and Na+ channels, implying that they are evolutionarily related (5).HVA channels are heteromeric complexes formed by the CaV α1 polypeptide, with several accessory subunits non-covalently bound. For example, the cardiac L-type voltage-gated calcium channel is formed by the CaV1.2 subunit in association with the soluble β-polypeptide localized to the intracellular side of the plasma membrane and a largely extracellular α2δ subunit (9, 10). The β-subunit has a role in regulating activation, inactivation, and voltage dependence as well as targeting of CaV1.2 to the plasma membrane. The crystal structure of the core region of the β-polypeptide in complex with a peptide corresponding to the interacting region of the CaV1.2 (AID) has been described (11, 12), revealing that it is comprised of two domains, a type 3 Src homology (SH3) domain and a guanylate kinase-like domain. CaV1.2 is associated, on the extracellular side of the membrane, with the α2δ subunit, the product of post-translational cleavage of a single gene comprised of a glycosylated extracellular α2 domain linked by disulfide bonds to the transmembrane δ polypeptide, which is also mainly extracellular and glycosylated. Four isoforms of α2δ have been identified (13, 14). The role of the α2δ protein is not as well understood as that of the β-subunit, but it has been shown to increase the current amplitude and have effects on inactivation (15). An additional membrane-spanning auxiliary subunit, γ, was initially thought to be unique to the skeletal muscle LTCC; however, recent studies have identified neuronal isoforms, although it remains unclear whether they have any role as calcium channel subunits (16, 17).We report here the purification of a recombinant CaV3.1, leading to the calculation of the first three-dimensional structure for a member of the LVA channel family. CaV3.1 is formed by two distinct segments, which we have been able to assign to the transmembrane and cytoplasmic domains. We have identified the C-terminal domain that forms a tail that winds around the base of the structure, providing insights as to how this channel may be regulated through the binding of accessory/regulatory proteins and/or long range conformational movements. Furthermore, we have been able to utilize this new three-dimensional structure to probe the assembly of the polypeptides forming the cardiac LTCCs after having also calculated a novel three-dimensional structure for the monomeric form of the channel purified from bovine heart. At the resolutions described here, CaV3.1 can be considered analogous to CaV1.2; see Fig. 1A for a sequence alignment overview. No mandatory auxiliary subunits for the T-type, LVA, channels have been identified. However, studies have shown that co-expression of CaV3.1 with α2δ subunits led to a 2-fold increase in T-type-mediated currents (18), and thus this model may also provide an insight as to how accessory proteins may associate with CaV3.1 to exert regulatory effects.Open in a separate windowFIGURE 1.Characterization, purification and image analysis of the T-type voltage gated calcium channel Cav3.1. A, schematic overview of a sequence comparison of voltage-gated calcium Ca2+ channel subunits CaV1.2 and CaV3.1. Sequence alignment was carried out using ClustalW (37). Solid regions indicate aligned sequences (black blocks correspond to the transmembrane helices); extracellular loops comprising <10 amino acids are not depicted. B, lane 1, silver-stained 10% SDS-PAGE of purified recombinant CaV3.1 revealing a single polypeptide band at ∼250 kDa. Lane 2, identification of the purified Cav3.1 by Western blotting (anti-CaV3.1, Santa Cruz Biotechnology sc-25690). Lane 3, Western blot of the purified Cav3.1 using an anti-His (Santa Cruz Biotechnology) antibody revealing a single protein product (recombinant Cav3.1 with a C-terminal His tag) at ∼250 kDa. C, field of negatively stained (2% w/v uranyl acetate) recombinant CaV3.1 showing white protein particles presenting a range of orientations ∼85–115 Å in size. The asterisk indicates a small area of aggregation that is easily distinguishable from the single CaV3.1 complexes. The black arrows indicate square-shaped particles with a side length of ∼100 Å. D, column I, examples of reference-free class averages obtained by alignment of the raw data that are representative of the range of multiple orientations sampled (box size 230 × 230 Å). Column II, corresponding back projections of the final three-dimensional volume illustrate that the structural features are consistent with those shown in the class averages. E, Fourier shell correlation plot indicating at a correlation of 0.5 that the three-dimensional CaV3.1 structure is at a resolution of 23 Å.  相似文献   

17.
Low voltage-activated T-type Cav3.2 calcium channels are expressed in neurosecretory chromaffin cells of the adrenal medulla. Previous studies have shown that naïve adrenal chromaffin cells express a nominal Cav3.2-dependent conductance. However, Cav3.2 conductance is up-regulated following chronic hypoxia or long term exposure to cAMP analogs. Thus, although a link between chronic stressors and up-regulation of Cav3.2 exists, there are no reports testing the specific role of Cav3.2 channels in the acute sympathoadrenal stress response. In this study, we examined the effects of acute sympathetic stress on T-type Cav3.2 calcium influx in mouse chromaffin cells in situ. Pituitary adenylate cyclase-activating peptide (PACAP) is an excitatory neuroactive peptide transmitter released by the splanchnic nerve under elevated sympathetic activity to stimulate the adrenal medulla. PACAP stimulation did not evoke action potential firing in chromaffin cells but did cause a persistent subthreshold membrane depolarization that resulted in an immediate and robust Ca2+-dependent catecholamine secretion. Moreover, PACAP-evoked secretion was sensitive to block by nickel chloride and was acutely inhibited by protein kinase C blockers. We utilized perforated patch electrophysiological recordings conducted in adrenal tissue slices to investigate the mechanism of PACAP-evoked calcium entry. We provide evidence that stimulation with exogenous PACAP and native neuronal stress stimulation both lead to a protein kinase C-mediated phosphodependent recruitment of a T-type Cav3.2 Ca2+ influx. This in turn evokes catecholamine release during the acute sympathetic stress response.  相似文献   

18.
Prior to maturation, mouse oocytes are arrested at the germinal vesicle (GV) stage during which they experience constitutive calcium (Ca2+) influx and spontaneous Ca2+ oscillations. The oscillations cease during maturation but Ca2+ influx continues, as the oocytes’ internal stores attain maximal content at the culmination of maturation, the metaphase II stage. The identity of the channel(s) that underlie this Ca2+ influx has not been completely determined. GV and matured oocytes are known to express three Ca2+ channels, CaV3.2, TRPV3 and TRPM7, but females null for each of these channels are fertile and their oocytes display minor modifications in Ca2+ homeostasis, suggesting a complex regulation of Ca2+ influx. To define the contribution of these channels at the GV stage, we used different divalent cations, pharmacological inhibitors and genetic models. We found that the three channels are active at this stage. CaV3.2 and TRPM7 channels contributed the majority of Ca2+ influx, as inhibitors and oocytes from homologous knockout (KO) lines showed severely reduced Ca2+ entry. Sr2+ influx was promoted by CaV3.2 channels, as Sr2+ oscillations were negligible in CaV3.2-KO oocytes but robust in control and Trpv3-KO GV oocytes. Mn2+ entry relied on expression of CaV3.2 and TRPM7 channels, but Ni2+ entry depended on the latter. CaV3.2 and TRPV3 channels combined to fill the Ca2+ stores, although CaV3.2 was the most impactful. Studies with pharmacological inhibitors effectively blocked the influx of divalent cations, but displayed off-target effects, and occasionally agonist-like properties. In conclusion, GV oocytes express channels mediating Ca2+ and other divalent cation influx that are pivotal for fertilization and early development. These channels may serve as targets for intervention to improve the success of assisted reproductive technologies.  相似文献   

19.
Cav3.2 T-type channels contain a high affinity metal binding site for trace metals such as copper and zinc. This site is occupied at physiologically relevant concentrations of these metals, leading to decreased channel activity and pain transmission. A histidine at position 191 was recently identified as a critical determinant for both trace metal block of Cav3.2 and modulation by redox agents. His191 is found on the extracellular face of the Cav3.2 channel on the IS3-S4 linker and is not conserved in other Cav3 channels. Mutation of the corresponding residue in Cav3.1 to histidine, Gln172, significantly enhances trace metal inhibition, but not to the level observed in wild-type Cav3.2, implying that other residues also contribute to the metal binding site. The goal of the present study is to identify these other residues using a series of chimeric channels. The key findings of the study are that the metal binding site is composed of a Asp-Gly-His motif in IS3–S4 and a second aspartate residue in IS2. These results suggest that metal binding stabilizes the closed conformation of the voltage-sensor paddle in repeat I, and thereby inhibits channel opening. These studies provide insight into the structure of T-type channels, and identify an extracellular motif that could be targeted for drug development.  相似文献   

20.
T-type Ca2+ channels have been implicated in tremorogenesis and motor coordination. The α1 subunit of the CaV3.1 T-type Ca2+ channel is highly expressed in motor pathways in the brain, but knockout of the CaV3.1 gene (α1G-/-) per se causes no motor defects in mice. Thus, the role of CaV3.1 channels in motor control remains obscure in vivo. Here, we investigated the effect of the CaV3.1 knockout in the null genetic background of α1 GABAA receptor (α1−/−) by generating the double mutants (α1−/−/α1G-/-). α1−/−/α1G-/- mice showed severer motor abnormalities than α1−/− mice as measured by potentiated tremor activities at 20 Hz and impaired motor learning. Propranolol, an anti-ET drug that is known to reduce the pathologic tremor in α1−/− mice, was not effective for suppressing the potentiated tremor in α1−/−/α1G-/- mice. In addition, α1−/−/α1G-/- mice showed an age-dependent loss of cerebellar Purkinje neurons. These results suggest that α1−/−/α1G-/- mice are a novel mouse model for a distinct subtype of ET in human and that CaV3.1 T-type Ca2+ channels play a role in motor coordination under pathological conditions.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号