Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 212-217, 2024 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-39097871

RESUMO

Due to the widespread use of methamphetamine (METH) among reproductive-aged women, the effects of intrauterine exposure to METH need to be investigated, as previous studies on this topic have been limited. The goal of this study is to examine the influence of two regulatory genes (miRNA-151-3p and CACNA1C) on the intrauterine life of mice exposed to METH. Pregnant mice received doses of 2 and 5 mg/kg of METH and saline from day 10 of pregnancy until the end. Their offspring were then evaluated for miRNA-151-3p and CACNA1C gene expression levels using real-time PCR. The findings indicated that exposure to METH reduced the expression levels of both miRNA-151-3p and CACNA1C genes in offspring compared to the control group (p≤0.001). In conclusion, intrauterine exposure to METH leads to a decrease in expression levels of both miRNA-151-3p and CACNA1C genes, potentially disrupting regulatory pathways involving these genes and having an impact on male reproductive health.


Assuntos
Canais de Cálcio Tipo L , Regulação para Baixo , Metanfetamina , MicroRNAs , Efeitos Tardios da Exposição Pré-Natal , Testículo , Animais , Metanfetamina/toxicidade , MicroRNAs/genética , MicroRNAs/metabolismo , Feminino , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Testículo/efeitos dos fármacos , Testículo/metabolismo , Ratos , Camundongos
2.
Nat Commun ; 15(1): 6649, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39103356

RESUMO

Vasodilation in response to low oxygen (O2) tension (hypoxic vasodilation) is an essential homeostatic response of systemic arteries that facilitates O2 supply to tissues according to demand. However, how blood vessels react to O2 deficiency is not well understood. A common belief is that arterial myocytes are O2-sensitive. Supporting this concept, it has been shown that the activity of myocyte L-type Ca2+channels, the main ion channels responsible for vascular contractility, is reversibly inhibited by hypoxia, although the underlying molecular mechanisms have remained elusive. Here, we show that genetic or pharmacological disruption of mitochondrial electron transport selectively abolishes O2 modulation of Ca2+ channels and hypoxic vasodilation. Mitochondria function as O2 sensors and effectors that signal myocyte Ca2+ channels due to constitutive Hif1α-mediated expression of specific electron transport subunit isoforms. These findings reveal the acute O2-sensing mechanisms of vascular cells and may guide new developments in vascular pharmacology.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia , Mitocôndrias , Oxigênio , Vasodilatação , Animais , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Transdução de Sinais , Masculino , Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Artérias/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Camundongos Knockout , Transporte de Elétrons , Canais de Cálcio/metabolismo , Canais de Cálcio/genética
3.
PLoS One ; 19(7): e0305248, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38968219

RESUMO

Long QT Syndrome type 8 (LQT8) is a cardiac arrhythmic disorder associated with Timothy Syndrome, stemming from mutations in the CACNA1C gene, particularly the G406R mutation. While prior studies hint at CACNA1C mutations' role in ventricular arrhythmia genesis, the mechanisms, especially in G406R presence, are not fully understood. This computational study explores how the G406R mutation, causing increased transmural dispersion of repolarization, induces and sustains reentrant ventricular arrhythmias. Using three-dimensional numerical simulations on an idealized left-ventricular model, integrating the Bidomain equations with the ten Tusscher-Panfilov ionic model, we observe that G406R mutation with 11% and 50% heterozygosis significantly increases transmural dispersion of repolarization. During S1-S4 stimulation protocols, these gradients facilitate conduction blocks, triggering reentrant ventricular tachycardia. Sustained reentry pathways occur only with G406R mutation at 50% heterozygosis, while neglecting transmural heterogeneities of action potential duration prevents stable reentry, regardless of G406R mutation presence.


Assuntos
Potenciais de Ação , Canais de Cálcio Tipo L , Simulação por Computador , Síndrome do QT Longo , Sindactilia , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Canais de Cálcio Tipo L/genética , Sindactilia/genética , Sindactilia/fisiopatologia , Mutação , Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Ventrículos do Coração/fisiopatologia , Modelos Cardiovasculares , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatologia
4.
Cell Rep ; 43(7): 114428, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38996073

RESUMO

To achieve the functional polarization that underlies brain computation, neurons sort protein material into distinct compartments. Ion channel composition, for example, differs between axons and dendrites, but the molecular determinants for their polarized trafficking remain obscure. Here, we identify mechanisms that target voltage-gated Ca2+ channels (CaVs) to distinct subcellular compartments. In hippocampal neurons, CaV2s trigger neurotransmitter release at the presynaptic active zone, and CaV1s localize somatodendritically. After knockout of all three CaV2s, expression of CaV2.1, but not CaV1.3, restores neurotransmitter release. We find that chimeric CaV1.3s with CaV2.1 intracellular C-termini localize to the active zone, mediate synaptic vesicle exocytosis, and render release sensitive to CaV1 blockers. This dominant targeting function of the CaV2.1 C-terminus requires the first EF hand in its proximal segment, and replacement of the CaV2.1 C-terminus with that of CaV1.3 abolishes CaV2.1 active zone localization and function. We conclude that CaV intracellular C-termini mediate compartment-specific targeting.


Assuntos
Hipocampo , Animais , Hipocampo/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo N/genética , Neurônios/metabolismo , Humanos , Camundongos , Ratos , Vesículas Sinápticas/metabolismo , Exocitose , Células HEK293
5.
Int J Mol Sci ; 25(14)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-39062812

RESUMO

Dietary intake of omega-3 polyunsaturated fatty acids (eicosapentaenoic acid, EPA) exerts antiarrhythmic effects, although the mechanisms are poorly understood. Here, we investigated the possible beneficial actions of EPA on saturated fatty acid-induced changes in the L-type Ca2+ channel in cardiomyocytes. Cardiomyocytes were cultured with an oleic acid/palmitic acid mixture (OAPA) in the presence or absence of EPA. Beating rate reduction in cardiomyocytes caused by OAPA were reversed by EPA. EPA also retrieved a reduction in Cav1.2 L-type Ca2+ current, mRNA, and protein caused by OAPA. Immunocytochemical analysis revealed a distinct downregulation of the Cav1.2 channel caused by OAPA with a concomitant decrease in the phosphorylated component of a transcription factor adenosine-3',5'-cyclic monophosphate (cAMP) response element binding protein (CREB) in the nucleus, which were rescued by EPA. A free fatty acid receptor 4 (FFAR4) agonist TUG-891 reversed expression of Cav1.2 and CREB mRNA caused by OAPA, whereas an FFAR4 antagonist AH-7614 abolished the effects of EPA. Excessive reactive oxygen species (ROS) accumulation caused by OAPA decreased Cav1.2 and CREB mRNA expressions, which was reversed by an ROS scavenger. Our data suggest that EPA rescues cellular Cav1.2-Ca2+ channel decline caused by OAPA lipotoxicity and oxidative stresses via both free fatty acid receptor 4-dependent and -independent pathways.


Assuntos
Canais de Cálcio Tipo L , Ácido Eicosapentaenoico , Miócitos Cardíacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Ácido Eicosapentaenoico/farmacologia , Animais , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Ratos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Ácidos Graxos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Cultivadas
6.
J Gen Physiol ; 156(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-38990175

RESUMO

L-type CaV1.2 current (ICa,L) links electrical excitation to contraction in cardiac myocytes. ICa,L is tightly regulated to control cardiac output. Rad is a Ras-related, monomeric protein that binds to L-type calcium channel ß subunits (CaVß) to promote inhibition of ICa,L. In addition to CaVß interaction conferred by the Rad core motif, the highly conserved Rad C-terminus can direct membrane association in vitro and inhibition of ICa,L in immortalized cell lines. In this work, we test the hypothesis that in cardiomyocytes the polybasic C-terminus of Rad confers t-tubular localization, and that membrane targeting is required for Rad-dependent ICa,L regulation. We introduced a 3xFlag epitope to the N-terminus of the endogenous mouse Rrad gene to facilitate analysis of subcellular localization. Full-length 3xFlag-Rad (Flag-Rad) mice were compared with a second transgenic mouse model, in which the extended polybasic C-termini of 3xFlag-Rad was truncated at alanine 277 (Flag-RadΔCT). Ventricular cardiomyocytes were isolated for anti-Flag-Rad immunocytochemistry and ex vivo electrophysiology. Full-length Flag-Rad showed a repeating t-tubular pattern whereas Flag-RadΔCT failed to display membrane association. ICa,L in Flag-RadΔCT cardiomyocytes showed a hyperpolarized activation midpoint and an increase in maximal conductance. Additionally, current decay was faster in Flag-RadΔCT cells. Myocardial ICa,L in a Rad C-terminal deletion model phenocopies ICa,L modulated in response to ß-AR stimulation. Mechanistically, the polybasic Rad C-terminus confers CaV1.2 regulation via membrane association. Interfering with Rad membrane association constitutes a specific target for boosting heart function as a treatment for heart failure with reduced ejection fraction.


Assuntos
Canais de Cálcio Tipo L , Miócitos Cardíacos , Animais , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Camundongos , Miócitos Cardíacos/metabolismo , Membrana Celular/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Camundongos Transgênicos , Proteínas ras
7.
J Pharmacol Sci ; 156(1): 45-48, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39068034

RESUMO

The gene product of ocular albinism 1 (OA1)/G-protein-coupled receptor (GPR)143 is a receptor for L-3,4-dihydroxyphenylanine (l-DOPA), the most effective agent for Parkinson's disease. When overexpressed, human wild-type GPR143, but not its mutants, inhibits neurite outgrowth in PC12 cells. We investigated the downstream signaling pathway for GPR143-induced inhibition of neurite outgrowth. Nifedipine restored GPR143-induced neurite outgrowth inhibition to the level of control transfectant but did not affect outgrowth in GPR143-knockdown cells. Cilnidipine and flunarizine also suppressed the GPR143-induced inhibition, but their effects at higher concentrations still occurred even in GPR143-knockdown cells. These results suggest that GPR143 regulates neurite outgrowth via L-type calcium channel(s).


Assuntos
Canais de Cálcio Tipo L , Crescimento Neuronal , Nifedipino , Receptores Acoplados a Proteínas G , Células PC12 , Animais , Ratos , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Nifedipino/farmacologia , Crescimento Neuronal/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Humanos , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas do Olho/farmacologia , Flunarizina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Levodopa/farmacologia , Técnicas de Silenciamento de Genes , Neuritos/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Glicoproteínas de Membrana
8.
J Biol Chem ; 300(7): 107470, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38879012

RESUMO

Resistance to inhibitors of cholinesterases (ric-8 proteins) are involved in modulating G-protein function, but little is known of their potential physiological importance in the heart. In the present study, we assessed the role of resistance to inhibitors of cholinesterase 8b (Ric-8b) in determining cardiac contractile function. We developed a murine model in which it was possible to conditionally delete ric-8b in cardiac tissue in the adult animal after the addition of tamoxifen. Deletion of ric-8b led to severely reduced contractility as measured using echocardiography days after administration of tamoxifen. Histological analysis of the ventricular tissue showed highly variable myocyte size, prominent fibrosis, and an increase in cellular apoptosis. RNA sequencing revealed transcriptional remodeling in response to cardiac ric-8b deletion involving the extracellular matrix and inflammation. Phosphoproteomic analysis revealed substantial downregulation of phosphopeptides related to myosin light chain 2. At the cellular level, the deletion of ric-8b led to loss of activation of the L-type calcium channel through the ß-adrenergic pathways. Using fluorescence resonance energy transfer-based assays, we showed ric-8b protein selectively interacts with the stimulatory G-protein, Gαs. We explored if deletion of Gnas (the gene encoding Gαs) in cardiac tissue using a similar approach in the mouse led to an equivalent phenotype. The conditional deletion of the Gαs gene in the ventricle led to comparable effects on contractile function and cardiac histology. We conclude that ric-8b is essential to preserve cardiac contractile function likely through an interaction with the stimulatory G-protein and downstream phosphorylation of myosin light chain 2.


Assuntos
Contração Miocárdica , Animais , Camundongos , Contração Miocárdica/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Cadeias Leves de Miosina/genética , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/genética , Miocárdio/metabolismo , Miocárdio/patologia , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Humanos , Inibidores da Colinesterase/farmacologia , Masculino , Apoptose/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina
9.
Artigo em Inglês | MEDLINE | ID: mdl-38945384

RESUMO

Environmental endocrine disrupting chemical 4-tert-butylphenol (4-tBP), a widely-utilized surfactant in various industries, poses potential risks to aquatic organisms. Our previous sequencing results suggested that 4-tBP-induced common carp liver injury might be associated with Ca2+ signaling and autophagy. However, the intricate involvement of these pathways in 4-tBP-induced cytotoxic mechanisms remained unexplored. To bridge these knowledge gaps, this study focused on epithelioma papulosum cyprini (EPC) cells, a significant cell type in fish biology. Initial observations showed that 4-tBP induced a dose-dependent perturbation in Ca2+ levels. Further investigations, with siRNA and L-type Ca2+ channel agonist (BAYK8644), identified L-type calcium channel gene CACNA1D as a critical regulator of 4-tBP-induced Ca2+ overload. Predictive analysis using miRanda platform suggested a potential interaction between miR-363 and CACNA1D, which was subsequently verified through dual-luciferase reporter gene assays. We then established miR-363 mimic/inhibitor models, along with miR-363 and CACNA1D co-suppression models in EPC cells. Through TEM observation, immunofluorescence assay, Ca2+ staining, and qRT-PCR analysis, we evaluated the role of miR-363/CACNA1D axis in modulating the effects of 4-tBP on Ca2+ signaling and autophagy. Results showed that miR-363 inhibitor exacerbated 4-tBP-induced increase in CALM2, CAMKII, Calpain2, and p62 expression and also led to decrease in ATG5, ATG7, and LC3b expression. In contrast, miR-363 mimic notably alleviated these changes. Notably, siRNA CACNA1D effectively modulating miR-363 inhibitor's effect. Our study revealed that 4-tBP induced Ca2+ overload and subsequent autophagy impairment via miR-363/CACNA1D axis. These findings illuminated a profound understanding of molecular mechanisms underlying 4-tBP-induced cytotoxicity and spotlighted a potential therapeutic target.


Assuntos
Autofagia , Cálcio , Disruptores Endócrinos , MicroRNAs , Animais , Autofagia/efeitos dos fármacos , MicroRNAs/metabolismo , MicroRNAs/genética , Cálcio/metabolismo , Disruptores Endócrinos/toxicidade , Carpas/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Fenóis/toxicidade , Poluentes Químicos da Água/toxicidade , Proteínas de Peixes/metabolismo , Proteínas de Peixes/genética
10.
Neuromuscul Disord ; 41: 24-28, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38870649

RESUMO

Primary hypokalemic periodic paralysis (HypoPP) is a skeletal muscle channelopathy most commonly caused by pathogenic variants in the calcium channel gene, CACNA1S. HypoPP can present with attacks of paralysis and/or permanent muscle weakness. Previous studies have shown that patients with HypoPP can have impaired quality of life (QoL). In this cross-sectional study, we aimed to describe the QoL in patients with HypoPP caused by pathogenic variants in CACNA1S using The Individualized Neuromuscular Quality of Life (INQoL) questionnaire, a validated tool to measure the QoL of patients with neuromuscular diseases (higher score, worse QoL). We showed that muscle weakness and fatigue were the symptoms with the greatest impact on participants' lives and that "activities", in the life domain of the INQoL, was most affected by HypoPP. Furthermore, we showed that the total INQoL score increased with age. Low QoL was primarily driven by progressive permanent muscle weakness and not attacks of paralysis, although half of the participants reported that attacks of paralysis challenged their daily life. The results suggest that special attention should be given to muscle weakness and fatigue in patients with HypoPP.


Assuntos
Paralisia Periódica Hipopotassêmica , Debilidade Muscular , Qualidade de Vida , Humanos , Masculino , Paralisia Periódica Hipopotassêmica/genética , Feminino , Adulto , Estudos Transversais , Pessoa de Meia-Idade , Inquéritos e Questionários , Adulto Jovem , Fadiga , Canais de Cálcio Tipo L/genética , Adolescente , Idoso
11.
Channels (Austin) ; 18(1): 2338782, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38691022

RESUMO

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.


Assuntos
Canais de Cálcio Tipo L , Miócitos Cardíacos , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/química , Animais , Miócitos Cardíacos/metabolismo , Humanos , Células HEK293 , Ratos , Domínios Proteicos
12.
Channels (Austin) ; 18(1): 2355121, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38762910

RESUMO

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.


Assuntos
Canais de Cálcio Tipo L , Proteínas dos Microfilamentos , Animais , Humanos , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Miócitos Cardíacos/metabolismo , Proteínas dos Microfilamentos/metabolismo
13.
Brain Struct Funct ; 229(5): 1299-1315, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38720004

RESUMO

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.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Canais de Cálcio Tipo L , Epistasia Genética , Esquizofrenia , Humanos , Fator Neurotrófico Derivado do Encéfalo/genética , Esquizofrenia/genética , Esquizofrenia/patologia , Feminino , Masculino , Adulto , Canais de Cálcio Tipo L/genética , Pessoa de Meia-Idade , Encéfalo/patologia , Polimorfismo de Nucleotídeo Único , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Imageamento por Ressonância Magnética , Adulto Jovem , Proteínas Ligadas por GPI
14.
Per Med ; 21(3): 145-150, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38722226

RESUMO

Background: Statins are commonly used medications. Variants in SLCO1B1, CYP2C9, and ABCG2 are known predictors of muscle effects when taking statins. More exploratory genes include RYR1 and CACNA1S, which can also be associated with disease conditions. Methods: Patients with pathogenic/likely pathogenic variants in RYR1 or CACNA1S were identified through an elective genomic testing program. Through chart review, patients with a history of statin use were assessed for statin-associated muscle symptoms (SAMS) along with collection of demographics and other known risk factors for SAMS. Results: Of the 23 patients who had a pathogenic or likely pathogenic RYR1 or CACNA1S variant found, 12 had previous statin use; of these, SAMS were identified in four patients. Conclusion: These data contribute to previous literature suggesting patients with RYR1 variants may have an increased SAMS risk. Additional research will be helpful in further investigating this relationship and providing recommendations.


[Box: see text].


Assuntos
Canais de Cálcio Tipo L , Inibidores de Hidroximetilglutaril-CoA Redutases , Canal de Liberação de Cálcio do Receptor de Rianodina , Humanos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Masculino , Canais de Cálcio Tipo L/genética , Feminino , Pessoa de Meia-Idade , Idoso , Incidência , Doenças Musculares/genética , Doenças Musculares/induzido quimicamente , Adulto , Fatores de Risco , Canais de Cálcio/genética
15.
J Neuromuscul Dis ; 11(4): 871-875, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38788083

RESUMO

Dominant mutations in CACNA1S gene mainly causes hypokalemic periodic paralysis (PP)(hypoPP). A 68-year-old male proband developed a progressive proximal weakness from the age of 35. Muscle biopsy showed atrophic fibers with vacuoles containing tubular aggregates. Exome sequencing revealed a heterozygous p.R528H (c.1583G>A) mutation in the CACNA1S gene. CACNA1S-related HypoPP evolving to persistent myopathy in late adulthood is a well-known clinical condition. However, isolated progressive myopathy (without PP) was only exceptionally reported and never with an early onset. Reporting a case of early onset CACNA1S-related myopathy in a patient with no HypoPP we intend to alert clinicians to consider it in the differential diagnosis of younger adult-onset myopathies especially when featuring vacuolar changes.


Assuntos
Canais de Cálcio Tipo L , Mutação , Humanos , Masculino , Idoso , Canais de Cálcio Tipo L/genética , Doenças Musculares/genética , Vacúolos/patologia , Vacúolos/genética , Músculo Esquelético/patologia , Idade de Início , Paralisia Periódica Hipopotassêmica/genética
16.
Channels (Austin) ; 18(1): 2335469, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38564754

RESUMO

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.


Assuntos
Bloqueadores dos Canais de Cálcio , Canais de Cálcio Tipo L , Camundongos , Humanos , Animais , Isradipino/farmacologia , Isradipino/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Administração Oral
17.
Cell Mol Life Sci ; 81(1): 164, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38575795

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental , Angiopatias Diabéticas , Hiperglicemia , Animais , Ratos , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Constrição , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Angiopatias Diabéticas/metabolismo , Glucose/metabolismo , Hiperglicemia/genética , Hiperglicemia/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Ratos Sprague-Dawley
18.
Nat Commun ; 15(1): 3528, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664444

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Envelhecimento , Miocárdio , Proteínas do Tecido Nervoso , Canal de Liberação de Cálcio do Receptor de Rianodina , Proteínas Supressoras de Tumor , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Masculino , Envelhecimento/metabolismo , Camundongos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Miocárdio/metabolismo , Miocárdio/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Técnicas de Silenciamento de Genes , Endossomos/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Coração/fisiopatologia , Camundongos Endogâmicos C57BL , Humanos , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética , Sístole
19.
Artigo em Inglês | MEDLINE | ID: mdl-38657895

RESUMO

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.


Assuntos
Anquirinas , Transtorno Bipolar , Canais de Cálcio Tipo L , Polimorfismo de Nucleotídeo Único , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Anquirinas/genética , Atenção/fisiologia , Transtorno Bipolar/genética , Transtorno Bipolar/psicologia , Canais de Cálcio Tipo L/genética , Cognição , Disfunção Cognitiva/genética , Memória de Curto Prazo , Testes Neuropsicológicos , População do Leste Asiático/genética
20.
Arterioscler Thromb Vasc Biol ; 44(6): 1202-1221, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38602101

RESUMO

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.


Assuntos
Proteínas de Ancoragem à Quinase A , Canais de Cálcio Tipo L , Artérias Cerebrais , Modelos Animais de Doenças , Hipertensão , Músculo Liso Vascular , Miócitos de Músculo Liso , Ratos Endogâmicos SHR , Animais , Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Hipertensão/fisiopatologia , Hipertensão/metabolismo , Hipertensão/genética , Artérias Cerebrais/metabolismo , Artérias Cerebrais/fisiopatologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Masculino , Miócitos de Músculo Liso/metabolismo , Condicionamento Físico Animal/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-alfa/genética , Sinalização do Cálcio , Camundongos Endogâmicos C57BL , Camundongos , Ratos , Ratos Endogâmicos WKY , Angiotensina II , Pressão Sanguínea , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA