Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.549
Filter
1.
J Assoc Physicians India ; 72(4): 54-58, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38881084

ABSTRACT

Calcium channel blockers (CCBs) are widely used antihypertensive agents due to their effectiveness in reducing blood pressure (BP), along with their good tolerability and evidence of reducing hypertension (HTN)-related cardiovascular and renal diseases. Cilnidipine, a unique dihydropyridine calcium antagonist, exhibits potent inhibitory action on both N-type and L-type voltage-dependent calcium channels. With excellent oral absorption and a prolonged duration of action, it demonstrates a significant antihypertensive effect. It effectively reduces BP both systolic and diastolic while providing renal, neurological, and cardiovascular protection. Unlike L-type CCBs, cilnidipine does not increase pulse rates (PRs) and is associated with reduced occurrence of pedal edema. Cilnidipine is an effective treatment choice for individuals with mild to moderate essential HTN, whether it is administered alone or in conjunction with other treatment modalities.


Subject(s)
Antihypertensive Agents , Calcium Channel Blockers , Dihydropyridines , Hypertension , Dihydropyridines/therapeutic use , Dihydropyridines/pharmacology , Humans , Calcium Channel Blockers/therapeutic use , Hypertension/drug therapy , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/pharmacology , Calcium Channels, N-Type/drug effects , Blood Pressure/drug effects , Calcium Channels, L-Type
2.
Cells ; 13(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38891076

ABSTRACT

Pacemaking activity in substantia nigra dopaminergic neurons is generated by the coordinated activity of a variety of distinct somatodendritic voltage- and calcium-gated ion channels. We investigated whether these functional interactions could arise from a common localization in macromolecular complexes where physical proximity would allow for efficient interaction and co-regulations. For that purpose, we immunopurified six ion channel proteins involved in substantia nigra neuron autonomous firing to identify their molecular interactions. The ion channels chosen as bait were Cav1.2, Cav1.3, HCN2, HCN4, Kv4.3, and SK3 channel proteins, and the methods chosen to determine interactions were co-immunoprecipitation analyzed through immunoblot and mass spectrometry as well as proximity ligation assay. A macromolecular complex composed of Cav1.3, HCN, and SK3 channels was unraveled. In addition, novel potential interactions between SK3 channels and sclerosis tuberous complex (Tsc) proteins, inhibitors of mTOR, and between HCN4 channels and the pro-degenerative protein Sarm1 were uncovered. In order to demonstrate the presence of these molecular interactions in situ, we used proximity ligation assay (PLA) imaging on midbrain slices containing the substantia nigra, and we could ascertain the presence of these protein complexes specifically in substantia nigra dopaminergic neurons. Based on the complementary functional role of the ion channels in the macromolecular complex identified, these results suggest that such tight interactions could partly underly the robustness of pacemaking in dopaminergic neurons.


Subject(s)
Dopaminergic Neurons , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Mesencephalon , Proteomics , Small-Conductance Calcium-Activated Potassium Channels , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Proteomics/methods , Dopaminergic Neurons/metabolism , Animals , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Mesencephalon/metabolism , Humans , Calcium Channels, L-Type/metabolism , Mice , Substantia Nigra/metabolism
3.
J Am Heart Assoc ; 13(12): e033733, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38860414

ABSTRACT

BACKGROUND: Chronic sympathetic stimulation drives desensitization and downregulation of ß1 adrenergic receptor (ß1AR) in heart failure. We aim to explore the differential downregulation subcellular pools of ß1AR signaling in the heart. METHODS AND RESULTS: We applied chronic infusion of isoproterenol to induced cardiomyopathy in male C57BL/6J mice. We applied confocal and proximity ligation assay to examine ß1AR association with L-type calcium channel, ryanodine receptor 2, and SERCA2a ((Sarco)endoplasmic reticulum calcium ATPase 2a) and Förster resonance energy transfer-based biosensors to probe subcellular ß1AR-PKA (protein kinase A) signaling in ventricular myocytes. Chronic infusion of isoproterenol led to reduced ß1AR protein levels, receptor association with L-type calcium channel and ryanodine receptor 2 measured by proximity ligation (puncta/cell, 29.65 saline versus 14.17 isoproterenol, P<0.05), and receptor-induced PKA signaling at the plasma membrane (Förster resonance energy transfer, 28.9% saline versus 1.9% isoproterenol, P<0.05) and ryanodine receptor 2 complex (Förster resonance energy transfer, 30.2% saline versus 10.6% isoproterenol, P<0.05). However, the ß1AR association with SERCA2a was enhanced (puncta/cell, 51.4 saline versus 87.5 isoproterenol, P<0.05), and the receptor signal was minimally affected. The isoproterenol-infused hearts displayed decreased PDE4D (phosphodiesterase 4D) and PDE3A and increased PDE2A, PDE4A, and PDE4B protein levels. We observed a reduced role of PDE4 and enhanced roles of PDE2 and PDE3 on the ß1AR-PKA activity at the ryanodine receptor 2 complexes and myocyte shortening. Despite the enhanced ß1AR association with SERCA2a, the endogenous norepinephrine-induced signaling was reduced at the SERCA2a complexes. Inhibiting monoamine oxidase A rescued the norepinephrine-induced PKA signaling at the SERCA2a and myocyte shortening. CONCLUSIONS: This study reveals distinct mechanisms for the downregulation of subcellular ß1AR signaling in the heart under chronic adrenergic stimulation.


Subject(s)
Calcium Channels, L-Type , Cyclic AMP-Dependent Protein Kinases , Down-Regulation , Isoproterenol , Mice, Inbred C57BL , Myocytes, Cardiac , Receptors, Adrenergic, beta-1 , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Signal Transduction , Animals , Receptors, Adrenergic, beta-1/metabolism , Male , Ryanodine Receptor Calcium Release Channel/metabolism , Isoproterenol/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/drug effects , Disease Models, Animal , Mice , Heart Failure/metabolism , Heart Failure/chemically induced , Heart Failure/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/chemically induced , Fluorescence Resonance Energy Transfer
4.
J Am Heart Assoc ; 13(12): e034893, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38879447

ABSTRACT

BACKGROUND: Advanced atrioventricular block (AVB), that is, higher than second-degree Mobitz-1, is an abnormal finding in athletes. Despite intensive investigation, in several cases the pathogenesis remains unknown, but frequently pacemaker implantation is still indicated. Increasing evidence points to circulating anti-Ro/Sjögren syndrome-related antigen A (SSA) antibodies cross-reacting with L-type calcium channel and inhibiting the related current as an epidemiologically relevant and potentially reversible cause of isolated AVB in adults. The aim of the study was to determine the prevalence of anti-Ro/SSA-associated advanced AVBs in a large sample of young athletes. METHODS AND RESULTS: A total of 2536 consecutive athletes aged <40 years without a history of cardiac diseases/interventions were enrolled in a cross-sectional study. Resting and exercise electrocardiography was performed, and those presenting any AVB were further evaluated by 24-hour Holter ECG. Athletes with second-degree AVBs and their mothers underwent anti-Ro/SSA testing. Moreover, purified immunoglobulin G from subjects with anti-Ro/SSA-positive and anti-Ro/SSA-negative advanced AVB were tested on L-type calcium current and L-type-calcium channel expression using tSA201 cells. The global prevalence of advanced AVB in the overall sample was ≈0.1%, but the risk considerably increased (2%) when intensely trained postpubertal male subjects were selectively considered. While none of the athletes with advanced AVB showed heart abnormalities, in 100% of cases anti-Ro/SSA antibodies were detected. Ex vivo experiments showed that immunoglobulin G from anti-Ro/SSA-positive but not -negative subjects with advanced AVB acutely inhibit L-type calcium current and chronically downregulate L-type-calcium channel expression. CONCLUSIONS: Our study provides evidence that advanced AVB occurs in young athletes, in most cases associated with anti-Ro/SSA antibodies blocking L-type calcium channels. These findings may open new avenues for immunomodulating therapies to reduce the risk of life-threatening events in athletes, avoiding or delaying pacemaker implantation.


Subject(s)
Antibodies, Antinuclear , Athletes , Atrioventricular Block , Calcium Channels, L-Type , Humans , Male , Female , Adult , Cross-Sectional Studies , Atrioventricular Block/immunology , Atrioventricular Block/epidemiology , Atrioventricular Block/diagnosis , Prevalence , Young Adult , Calcium Channels, L-Type/immunology , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Adolescent , Electrocardiography, Ambulatory , Ribonucleoproteins/immunology
5.
Brain Struct Funct ; 229(5): 1299-1315, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38720004

ABSTRACT

The expression of Neuritin-1 (NRN1), a neurotrophic factor crucial for neurodevelopment and synaptic plasticity, is enhanced by the Brain Derived Neurotrophic Factor (BDNF). Although the receptor of NRN1 remains unclear, it is suggested that NRN1's activation of the insulin receptor (IR) pathway promotes the transcription of the calcium voltage-gated channel subunit alpha1 C (CACNA1C). These three genes have been independently associated with schizophrenia (SZ) risk, symptomatology, and brain differences. However, research on how they synergistically modulate these phenotypes is scarce. We aimed to study whether the genetic epistasis between these genes affects the risk and clinical presentation of the disorder via its effect on brain structure. First, we tested the epistatic effect of NRN1 and BDNF or CACNA1C on (i) the risk for SZ, (ii) clinical symptoms severity and functionality (onset, PANSS, CGI and GAF), and (iii) brain cortical structure (thickness, surface area and volume measures estimated using FreeSurfer) in a sample of 86 SZ patients and 89 healthy subjects. Second, we explored whether those brain clusters influenced by epistatic effects mediate the clinical profiles. Although we did not find a direct epistatic impact on the risk, our data unveiled significant effects on the disorder's clinical presentation. Specifically, the NRN1-rs10484320 x BDNF-rs6265 interplay influenced PANSS general psychopathology, and the NRN1-rs4960155 x CACNA1C-rs1006737 interaction affected GAF scores. Moreover, several interactions between NRN1 SNPs and BDNF-rs6265 significantly influenced the surface area and cortical volume of the frontal, parietal, and temporal brain regions within patients. The NRN1-rs10484320 x BDNF-rs6265 epistasis in the left lateral orbitofrontal cortex fully mediated the effect on PANSS general psychopathology. Our study not only adds clinical significance to the well-described molecular relationship between NRN1 and BDNF but also underscores the utility of deconstructing SZ into biologically validated brain-imaging markers to explore their mediation role in the path from genetics to complex clinical manifestation.


Subject(s)
Brain-Derived Neurotrophic Factor , Calcium Channels, L-Type , Epistasis, Genetic , Schizophrenia , Humans , Brain-Derived Neurotrophic Factor/genetics , Schizophrenia/genetics , Schizophrenia/pathology , Female , Male , Adult , Calcium Channels, L-Type/genetics , Middle Aged , Brain/pathology , Polymorphism, Single Nucleotide , Neuropeptides/genetics , Neuropeptides/metabolism , Magnetic Resonance Imaging , Young Adult , GPI-Linked Proteins
6.
Molecules ; 29(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38792145

ABSTRACT

The Cupressaceae family includes species considered to be medicinal. Their essential oil is used for headaches, colds, cough, and bronchitis. Cedar trees like Chamaecyparis lawsoniana (C. lawsoniana) are commonly found in urban areas. We investigated whether C. lawsoniana exerts some of its effects by modifying airway smooth muscle (ASM) contractility. The leaves of C. lawsoniana (363 g) were pulverized mechanically, and extracts were obtained by successive maceration 1:10 (w:w) with methanol/CHCl3. Guinea pig tracheal rings were contracted with KCl, tetraethylammonium (TEA), histamine (HIS), or carbachol (Cch) in organ baths. In the Cch experiments, tissues were pre-incubated with D-600, an antagonist of L-type voltage-dependent Ca2+ channels (L-VDCC) before the addition of C. lawsoniana. Interestingly, at different concentrations, C. lawsoniana diminished the tracheal contractions induced by KCl, TEA, HIS, and Cch. In ASM cells, C. lawsoniana significantly diminished L-type Ca2+ currents. ASM cells stimulated with Cch produced a transient Ca2+ peak followed by a sustained plateau maintained by L-VDCC and store-operated Ca2+ channels (SOCC). C. lawsoniana almost abolished this last response. These results show that C. lawsoniana, and its active metabolite quercetin, relax the ASM by inhibiting the L-VDCC and SOCC; further studies must be performed to obtain the complete set of metabolites of the extract and study at length their pharmacological properties.


Subject(s)
Calcium , Chamaecyparis , Muscle Contraction , Muscle, Smooth , Plant Extracts , Quercetin , Trachea , Animals , Guinea Pigs , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle Contraction/drug effects , Quercetin/pharmacology , Quercetin/chemistry , Trachea/drug effects , Trachea/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Chamaecyparis/chemistry , Calcium/metabolism , Male , Calcium Channel Blockers/pharmacology , Histamine/metabolism , Calcium Channels, L-Type/metabolism , Plant Leaves/chemistry
7.
Commun Biol ; 7(1): 620, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783117

ABSTRACT

A key player of excitable cells in the heart and brain is the L-type calcium channel CaV1.3. In the heart, it is required for voltage-dependent Ca2+-signaling, i.e., for controlling and modulating atrial cardiomyocyte excitation-contraction coupling. The clustering of CaV1.3 in functionally relevant channel multimers has not been addressed due to a lack of stoichiometric labeling combined with high-resolution imaging. Here, we developed a HaloTag-labeling strategy to visualize and quantify CaV1.3 clusters using STED nanoscopy to address the questions of cluster size and intra-cluster channel density. Channel clusters were identified in the plasma membrane of transfected live HEK293 cells as well as in giant plasma membrane vesicles derived from these cells that were spread on modified glass support to obtain supported plasma membrane bilayers (SPMBs). A small fraction of the channel clusters was colocalized with early and recycling endosomes at the membranes. STED nanoscopy in conjunction with live-cell and SPMB imaging enabled us to quantify CaV1.3 cluster sizes and their molecular density revealing significantly lower channel densities than expected for dense channel packing. CaV1.3 channel cluster size and molecular density were increased in SPMBs after treatment of the cells with the sympathomimetic compound isoprenaline, suggesting a regulated channel cluster condensation mechanism.


Subject(s)
Calcium Channels, L-Type , Cell Membrane , Humans , HEK293 Cells , Cell Membrane/metabolism , Calcium Channels, L-Type/metabolism
8.
Mol Biol Cell ; 35(7): ar92, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38758660

ABSTRACT

Chromaffin cells of the adrenal medulla transduce sympathetic nerve activity into stress hormone secretion. The two neurotransmitters principally responsible for coupling cell stimulation to secretion are acetylcholine and pituitary adenylate activating polypeptide (PACAP). In contrast to acetylcholine, PACAP evokes a persistent secretory response from chromaffin cells. However, the mechanisms by which PACAP acts are poorly understood. Here, it is shown that PACAP induces sustained increases in cytosolic Ca2+ which are disrupted when Ca2+ influx through L-type channels is blocked or internal Ca2+ stores are depleted. PACAP liberates stored Ca2+ via inositol trisphosphate receptors (IP3Rs) on the endoplasmic reticulum (ER), thereby functionally coupling Ca2+ mobilization to Ca2+ influx and supporting Ca2+-induced Ca2+-release. These Ca2+ influx and mobilization pathways are unified by an absolute dependence on phospholipase C epsilon (PLCε) activity. Thus, the persistent secretory response that is a defining feature of PACAP activity, in situ, is regulated by a signaling network that promotes sustained elevations in intracellular Ca2+ through multiple pathways.


Subject(s)
Calcium Signaling , Calcium , Chromaffin Cells , Endoplasmic Reticulum , Inositol 1,4,5-Trisphosphate Receptors , Pituitary Adenylate Cyclase-Activating Polypeptide , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Chromaffin Cells/metabolism , Cattle , Calcium Channels, L-Type/metabolism
9.
Channels (Austin) ; 18(1): 2355121, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38762910

ABSTRACT

L-type calcium channels (LTCCs), the major portal for Ca2+ entry into cardiomyocytes, are essential for excitation-contraction coupling and thus play a central role in regulating overall cardiac function. LTCC function is finely tuned by multiple signaling pathways and accessory proteins. Leucine-rich repeat-containing protein 10 (LRRC10) is a little studied cardiomyocyte-specific protein recently identified as a modulator of LTCCs. LRRC10 exerts a remarkable effect on LTCC function, more than doubling L-type Ca2+ current (ICa,L) amplitude in a heterologous expression system by altering the gating of the channels without changing their surface membrane expression. Genetic ablation of LRRC10 expression in mouse and zebrafish hearts leads to a significant reduction in ICa,L density and a slowly progressive dilated cardiomyopathy in mice. Rare sequence variants of LRRC10 have been identified in dilated cardiomyopathy and sudden unexplained nocturnal cardiac death syndrome, but these variants have not been clearly linked to disease. Nevertheless, the DCM-associated variant, I195T, converted LRRC10 from a ICa,L potentiator to a ICa,L suppressor, thus illustrating the wide dynamic range of LRRC10-mediated ICa,L regulation. This review focuses on the contemporary knowledge of LTCC modulation by LRRC10 and discusses potential directions for future investigations.


Subject(s)
Calcium Channels, L-Type , Microfilament Proteins , Animals , Humans , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Myocytes, Cardiac/metabolism , Microfilament Proteins/metabolism
10.
Mol Cell Endocrinol ; 591: 112275, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38777212

ABSTRACT

Metabolic changes are critical in the regulation of Ca2+ influx in central and peripheral neuroendocrine cells. To study the regulation of L-type Ca2+ channels by AMPK we used biochemical reagents and ATP/glucose-concentration manipulations in rat chromaffin cells. AICAR and Compound-C, at low concentration, significantly induce changes in L-type Ca2+ channel-current amplitude and voltage dependence. Remarkably, an overlasting decrease in the channel-current density can be induced by lowering the intracellular level of ATP. Accordingly, Ca2+ channel-current density gradually diminishes by decreasing the extracellular glucose concentration. By using immunofluorescence, a decrease in the expression of CaV1.2 is observed while decreasing extracellular glucose, suggesting that AMPK reduces the number of functional Ca2+ channels into the plasma membrane. Together, these results support for the first time the dependence of metabolic changes in the maintenance of Ca2+ channel-current by AMPK. They reveal a key step in Ca2+ influx in secretory cells.


Subject(s)
AMP-Activated Protein Kinases , Aminoimidazole Carboxamide , Calcium Channels, L-Type , Chromaffin Cells , Glucose , Animals , Chromaffin Cells/metabolism , Chromaffin Cells/drug effects , Calcium Channels, L-Type/metabolism , AMP-Activated Protein Kinases/metabolism , Rats , Glucose/metabolism , Glucose/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Adenosine Triphosphate/metabolism , Ribonucleotides/pharmacology , Pyrimidines/pharmacology , Calcium/metabolism , Pyrazoles/pharmacology , Cells, Cultured , Rats, Wistar , Ion Channel Gating/drug effects
11.
Eur J Pharmacol ; 976: 176674, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38810715

ABSTRACT

Antipsychotic drugs often lead to adverse effects, including those related to the cardiovascular system. Of these, quetiapine is known to cause significant changes in the QT interval although the underlying mechanism remains mysterious, prompting us to examine its effects on cardiac electrophysiological properties. Therefore, we investigated the effect of quetiapine on contraction, action potential (AP), and the associated membrane currents such as L-type Ca2+ and K+ using the whole-cell patch clamp method to examine its impacts on isolated rat ventricular myocytes. Our results showed that (1) quetiapine reduces cell contractility in a concentration-dependent manner and (2) leads to a significant prolongation in the duration of AP in isolated ventricular myocytes. This effect was both concentration and frequency-dependent; (3) quetiapine significantly decreased the Ca2+, transient outward K+, and steady-state K+ currents. However, only high concentration of quetiapine (100 µM) could significantly change the activation and reactivation kinetics of L-type Ca2+ channels. This study demonstrates that QT extension induced by quetiapine is mainly associated with the prolongation of AP. Moreover, quetiapine caused a significant decrease in contractile force and excitability of ventricular myocytes by suppressing Ca2+ and K+ currents.


Subject(s)
Action Potentials , Calcium Channels, L-Type , Heart Ventricles , Myocardial Contraction , Myocytes, Cardiac , Quetiapine Fumarate , Animals , Quetiapine Fumarate/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Heart Ventricles/drug effects , Heart Ventricles/cytology , Rats , Action Potentials/drug effects , Male , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/drug effects , Myocardial Contraction/drug effects , Rats, Sprague-Dawley , Antipsychotic Agents/pharmacology , Dose-Response Relationship, Drug , Calcium/metabolism
12.
BMC Oral Health ; 24(1): 552, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735923

ABSTRACT

Patients who suffer from myofascial orofacial pain could affect their quality of life deeply. The pathogenesis of pain is still unclear. Our objective was to assess Whether Voltage-gated calcium channel α2δ-1(Cavα2δ-1) is related to myofascial orofacial pain. Rats were divided into the masseter tendon ligation group and the sham group. Compared with the sham group, the mechanical pain threshold of the masseter tendon ligation group was reduced on the 4th, 7th, 10th and 14th day after operation(P < 0.05). On the 14th day after operation, Cavα2δ-1 mRNA expression levels in trigeminal ganglion (TG) and the trigeminal spinal subnucleus caudalis and C1-C2 spinal cervical dorsal horn (Vc/C2) of the masseter tendon ligation group were increased (PTG=0.021, PVc/C2=0.012). Rats were divided into three groups. On the 4th day after ligating the superficial tendon of the left masseter muscle of the rats, 10 ul Cavα2δ-1 antisense oligonucleotide, 10 ul Cavα2δ-1 mismatched oligonucleotides and 10 ul normal saline was separately injected into the left masseter muscle of rats in Cavα2δ-1 antisense oligonucleotide group, Cavα2δ-1 mismatched oligonucleotides group and normal saline control group twice a day for 4 days. The mechanical pain threshold of the Cavα2δ-1 antisense oligonucleotides group was higher than Cavα2δ-1 mismatched oligonucleotides group on the 7th and 10th day after operation (P < 0.01). After PC12 cells were treated with lipopolysaccharide, Cavα2δ-1 mRNA expression level increased (P < 0.001). Cavα2δ-1 may be involved in the occurrence and development in myofascial orofacial pain.


Subject(s)
Calcium Channels , Masseter Muscle , Rats, Sprague-Dawley , Trigeminal Ganglion , Animals , Rats , Masseter Muscle/metabolism , Male , Calcium Channels/metabolism , Trigeminal Ganglion/metabolism , Pain Threshold , Facial Pain/metabolism , Spinal Cord Dorsal Horn/metabolism , Oligonucleotides, Antisense/pharmacology , Myofascial Pain Syndromes , RNA, Messenger/metabolism , Calcium Channels, L-Type
13.
Channels (Austin) ; 18(1): 2338782, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38691022

ABSTRACT

L-type calcium channels are essential for the excitation-contraction coupling in cardiac muscle. The CaV1.2 channel is the most predominant isoform in the ventricle which consists of a multi-subunit membrane complex that includes the CaV1.2 pore-forming subunit and auxiliary subunits like CaVα2δ and CaVß2b. The CaV1.2 channel's C-terminus undergoes proteolytic cleavage, and the distal C-terminal domain (DCtermD) associates with the channel core through two domains known as proximal and distal C-terminal regulatory domain (PCRD and DCRD, respectively). The interaction between the DCtermD and the remaining C-terminus reduces the channel activity and modifies voltage- and calcium-dependent inactivation mechanisms, leading to an autoinhibitory effect. In this study, we investigate how the interaction between DCRD and PCRD affects the inactivation processes and CaV1.2 activity. We expressed a 14-amino acid peptide miming the DCRD-PCRD interaction sequence in both heterologous systems and cardiomyocytes. Our results show that overexpression of this small peptide can displace the DCtermD and replicate the effects of the entire DCtermD on voltage-dependent inactivation and channel inhibition. However, the effect on calcium-dependent inactivation requires the full DCtermD and is prevented by overexpression of calmodulin. In conclusion, our results suggest that the interaction between DCRD and PCRD is sufficient to bring about the current inhibition and alter the voltage-dependent inactivation, possibly in an allosteric manner. Additionally, our data suggest that the DCtermD competitively modifies the calcium-dependent mechanism. The identified peptide sequence provides a valuable tool for further dissecting the molecular mechanisms that regulate L-type calcium channels' basal activity in cardiomyocytes.


Subject(s)
Calcium Channels, L-Type , Myocytes, Cardiac , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/chemistry , Animals , Myocytes, Cardiac/metabolism , Humans , HEK293 Cells , Rats , Protein Domains
14.
Nature ; 628(8009): 818-825, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38658687

ABSTRACT

Timothy syndrome (TS) is a severe, multisystem disorder characterized by autism, epilepsy, long-QT syndrome and other neuropsychiatric conditions1. TS type 1 (TS1) is caused by a gain-of-function variant in the alternatively spliced and developmentally enriched CACNA1C exon 8A, as opposed to its counterpart exon 8. We previously uncovered several phenotypes in neurons derived from patients with TS1, including delayed channel inactivation, prolonged depolarization-induced calcium rise, impaired interneuron migration, activity-dependent dendrite retraction and an unanticipated persistent expression of exon 8A2-6. We reasoned that switching CACNA1C exon utilization from 8A to 8 would represent a potential therapeutic strategy. Here we developed antisense oligonucleotides (ASOs) to effectively decrease the inclusion of exon 8A in human cells both in vitro and, following transplantation, in vivo. We discovered that the ASO-mediated switch from exon 8A to 8 robustly rescued defects in patient-derived cortical organoids and migration in forebrain assembloids. Leveraging a transplantation platform previously developed7, we found that a single intrathecal ASO administration rescued calcium changes and in vivo dendrite retraction of patient neurons, suggesting that suppression of CACNA1C exon 8A expression is a potential treatment for TS1. Broadly, these experiments illustrate how a multilevel, in vivo and in vitro stem cell model-based approach can identify strategies to reverse disease-relevant neural pathophysiology.


Subject(s)
Autistic Disorder , Long QT Syndrome , Oligonucleotides, Antisense , Syndactyly , Animals , Female , Humans , Male , Mice , Alternative Splicing/drug effects , Alternative Splicing/genetics , Autistic Disorder/drug therapy , Autistic Disorder/genetics , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Cell Movement/drug effects , Dendrites/metabolism , Exons/genetics , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Neurons/metabolism , Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Organoids/drug effects , Organoids/metabolism , Prosencephalon/metabolism , Prosencephalon/cytology , Syndactyly/drug therapy , Syndactyly/genetics , Interneurons/cytology , Interneurons/drug effects
15.
Nat Commun ; 15(1): 3528, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664444

ABSTRACT

Cardiac dysfunction is a hallmark of aging in humans and mice. Here we report that a two-week treatment to restore youthful Bridging Integrator 1 (BIN1) levels in the hearts of 24-month-old mice rejuvenates cardiac function and substantially reverses the aging phenotype. Our data indicate that age-associated overexpression of BIN1 occurs alongside dysregulated endosomal recycling and disrupted trafficking of cardiac CaV1.2 and type 2 ryanodine receptors. These deficiencies affect channel function at rest and their upregulation during acute stress. In vivo echocardiography reveals reduced systolic function in old mice. BIN1 knockdown using an adeno-associated virus serotype 9 packaged shRNA-mBIN1 restores the nanoscale distribution and clustering plasticity of ryanodine receptors and recovers Ca2+ transient amplitudes and cardiac systolic function toward youthful levels. Enhanced systolic function correlates with increased phosphorylation of the myofilament protein cardiac myosin binding protein-C. These results reveal BIN1 knockdown as a novel therapeutic strategy to rejuvenate the aging myocardium.


Subject(s)
Adaptor Proteins, Signal Transducing , Aging , Myocardium , Nerve Tissue Proteins , Ryanodine Receptor Calcium Release Channel , Tumor Suppressor Proteins , Animals , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Male , Aging/metabolism , Mice , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Myocardium/metabolism , Myocardium/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Gene Knockdown Techniques , Endosomes/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Heart/physiopathology , Mice, Inbred C57BL , Humans , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Systole
16.
Arterioscler Thromb Vasc Biol ; 44(6): 1202-1221, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38602101

ABSTRACT

BACKGROUND: Hypertension is a major, prevalent risk factor for the development and progression of cerebrovascular disease. Regular exercise has been recommended as an excellent choice for the large population of individuals with mild-to-moderate elevations in blood pressure, but the mechanisms that underlie its vascular-protective and antihypertensive effects remain unknown. Here, we describe a mechanism by which myocyte AKAP150 (A-kinase anchoring protein 150) inhibition induced by exercise training alleviates voltage-dependent L-type Ca2+ channel (CaV1.2) activity and restores cerebral arterial function in hypertension. METHODS: Spontaneously hypertensive rats and newly generated smooth muscle-specific AKAP150 knockin mice were used to assess the role of myocyte AKAP150/CaV1.2 channel in regulating cerebral artery function after exercise intervention. RESULTS: Activation of the AKAP150/PKCα (protein kinase Cα) signaling increased CaV1.2 activity and Ca2+ influx of cerebral arterial myocyte, thus enhancing vascular tone in spontaneously hypertensive rats. Smooth muscle-specific AKAP150 knockin mice were hypertensive with higher CaV1.2 channel activity and increased vascular tone. Furthermore, treatment of Ang II (angiotensin II) resulted in a more pronounced increase in blood pressure in smooth muscle-specific AKAP150 knockin mice. Exercise training significantly reduced arterial myocyte AKAP150 expression and alleviated CaV1.2 channel activity, thus restoring cerebral arterial function in spontaneously hypertensive rats and smooth muscle-specific AKAP150 knockin mice. AT1R (AT1 receptor) and AKAP150 were interacted closely in arterial myocytes. Exercise decreased the circulating Ang II and Ang II-involved AT1R-AKAP150 association in myocytes of hypertension. CONCLUSIONS: The current study demonstrates that aerobic exercise ameliorates CaV1.2 channel function via inhibiting myocyte AKAP150, which contributes to reduced cerebral arterial tone in hypertension.


Subject(s)
A Kinase Anchor Proteins , Calcium Channels, L-Type , Cerebral Arteries , Disease Models, Animal , Hypertension , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Rats, Inbred SHR , Animals , A Kinase Anchor Proteins/metabolism , A Kinase Anchor Proteins/genetics , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Hypertension/physiopathology , Hypertension/metabolism , Hypertension/genetics , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Male , Myocytes, Smooth Muscle/metabolism , Physical Conditioning, Animal/physiology , Protein Kinase C-alpha/metabolism , Protein Kinase C-alpha/genetics , Calcium Signaling , Mice, Inbred C57BL , Mice , Rats , Rats, Inbred WKY , Angiotensin II , Blood Pressure , Signal Transduction
17.
Article in English | MEDLINE | ID: mdl-38657895

ABSTRACT

Bipolar disorder (BD) is a complex, severe mental illness with cognitive impairment. Impairments in attention and memory are particularly evident. A large number of previous studies have identified CACNA1C and ANK3 gene variants as risk factors for BD and both affect cognitive function in people with BD. However, it is unclear whether there is an interaction effects between the two genes on cognitive impairment in patients. We used 153 Chinese Han Chinese patients with BD to explore the association of CACNA1C and ANK3 variants with attention and immediate memory using Plink software and and performed a epistatic interaction effects analysis. We found that CACNA1C and ANK3 gene variants respectively affected patients' scores on attention and memory tests. The significant SNP in the CACNA1C and ANK3 genes are rs73042126(P = 3.16 × 10-5,FDR = 0.0253) and rs2393640(P = 1.50 × 10-4,FDR = 0.0353) respectively. And they also interacted to affect cognitive functioning in BD patients (attention: P = 0.0289; immediate memory: P = 0.0398). Follow-up studies should increase the sample size, improve the assessment methods and experimental design, and further explore the pathogenic mechanisms of BD.


Subject(s)
Ankyrins , Bipolar Disorder , Calcium Channels, L-Type , Polymorphism, Single Nucleotide , Adult , Female , Humans , Male , Middle Aged , Ankyrins/genetics , Attention/physiology , Bipolar Disorder/genetics , Bipolar Disorder/psychology , Calcium Channels, L-Type/genetics , Cognition , Cognitive Dysfunction/genetics , Memory, Short-Term , Neuropsychological Tests , East Asian People/genetics
18.
Channels (Austin) ; 18(1): 2341077, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38601983

ABSTRACT

Voltage-gated calcium channels (VGCCs) are the major conduits for calcium ions (Ca2+) within excitable cells. Recent studies have highlighted the non-ionotropic functionality of VGCCs, revealing their capacity to activate intracellular pathways independently of ion flow. This non-ionotropic signaling mode plays a pivotal role in excitation-coupling processes, including gene transcription through excitation-transcription (ET), synaptic transmission via excitation-secretion (ES), and cardiac contraction through excitation-contraction (EC). However, it is noteworthy that these excitation-coupling processes require extracellular calcium (Ca2+) and Ca2+ occupancy of the channel ion pore. Analogous to the "non-canonical" characterization of the non-ionotropic signaling exhibited by the N-methyl-D-aspartate receptor (NMDA), which requires extracellular Ca2+ without the influx of ions, VGCC activation requires depolarization-triggered conformational change(s) concomitant with Ca2+ binding to the open channel. Here, we discuss the contributions of VGCCs to ES, ET, and EC coupling as Ca2+ binding macromolecules that transduces external stimuli to intracellular input prior to elevating intracellular Ca2+. We emphasize the recognition of calcium ion occupancy within the open ion-pore and its contribution to the excitation coupling processes that precede the influx of calcium. The non-ionotropic activation of VGCCs, triggered by the upstroke of an action potential, provides a conceptual framework to elucidate the mechanistic aspects underlying the microseconds nature of synaptic transmission, cardiac contractility, and the rapid induction of first-wave genes.


Subject(s)
Calcium Channels , Calcium , Calcium/metabolism , Calcium Channels/metabolism , Signal Transduction , Excitation Contraction Coupling , Ions/metabolism , Calcium Signaling/physiology , Calcium Channels, L-Type/metabolism
19.
Channels (Austin) ; 18(1): 2335469, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38564754

ABSTRACT

Studies in genetically modified animals and human genetics have recently provided new insight into the role of voltage-gated L-type Ca2+ channels in human disease. Therefore, the inhibition of L-type Ca2+ channels in vivo in wildtype and mutant mice by potent dihydropyridine (DHP) Ca2+ channel blockers serves as an important pharmacological tool. These drugs have a short plasma half-life in humans and especially in rodents and show high first-pass metabolism upon oral application. In the vast majority of in vivo studies, they have therefore been delivered through parenteral routes, mostly subcutaneously or intraperitoneally. High peak plasma concentrations of DHPs cause side effects, evident as DHP-induced aversive behaviors confounding the interpretation of behavioral readouts. Nevertheless, pharmacokinetic data measuring the exposure achieved with these applications are sparse. Moreover, parenteral injections require animal handling and can be associated with pain, discomfort and stress which could influence a variety of physiological processes, behavioral and other functional readouts. Here, we describe a noninvasive oral application of the DHP isradipine by training mice to quickly consume small volumes of flavored yogurt that can serve as drug vehicle. This procedure does not require animal handling, allows repeated drug application over several days and reproducibly achieves peak plasma concentrations over a wide range previously shown to be well-tolerated in humans. This protocol should facilitate ongoing nonclinical studies in mice exploring new indications for DHP Ca2+ channel blockers.


Subject(s)
Calcium Channel Blockers , Calcium Channels, L-Type , Mice , Humans , Animals , Isradipine/pharmacology , Isradipine/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Administration, Oral
20.
Cell Mol Life Sci ; 81(1): 164, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575795

ABSTRACT

Diabetic hyperglycemia induces dysfunctions of arterial smooth muscle, leading to diabetic vascular complications. The CaV1.2 calcium channel is one primary pathway for Ca2+ influx, which initiates vasoconstriction. However, the long-term regulation mechanism(s) for vascular CaV1.2 functions under hyperglycemic condition remains unknown. Here, Sprague-Dawley rats fed with high-fat diet in combination with low dose streptozotocin and Goto-Kakizaki (GK) rats were used as diabetic models. Isolated mesenteric arteries (MAs) and vascular smooth muscle cells (VSMCs) from rat models were used to assess K+-induced arterial constriction and CaV1.2 channel functions using vascular myograph and whole-cell patch clamp, respectively. K+-induced vasoconstriction is persistently enhanced in the MAs from diabetic rats, and CaV1.2 alternative spliced exon 9* is increased, while exon 33 is decreased in rat diabetic arteries. Furthermore, CaV1.2 channels exhibit hyperpolarized current-voltage and activation curve in VSMCs from diabetic rats, which facilitates the channel function. Unexpectedly, the application of glycated serum (GS), mimicking advanced glycation end-products (AGEs), but not glucose, downregulates the expression of the splicing factor Rbfox1 in VSMCs. Moreover, GS application or Rbfox1 knockdown dynamically regulates alternative exons 9* and 33, leading to facilitated functions of CaV1.2 channels in VSMCs and MAs. Notably, GS increases K+-induced intracellular calcium concentration of VSMCs and the vasoconstriction of MAs. These results reveal that AGEs, not glucose, long-termly regulates CaV1.2 alternative splicing events by decreasing Rbfox1 expression, thereby enhancing channel functions and increasing vasoconstriction under diabetic hyperglycemia. This study identifies the specific molecular mechanism for enhanced vasoconstriction under hyperglycemia, providing a potential target for managing diabetic vascular complications.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Angiopathies , Hyperglycemia , Animals , Rats , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Constriction , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/metabolism , Glucose/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...