Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Heart Rhythm ; 18(10): 1760-1771, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34023501

RESUMO

BACKGROUND: Atrial fibrillation (AF), a common arrhythmia in clinics, is characterized as downregulation of L-type calcium channel (LTCC) and shortening of atrial action potential duration (APD). Our prior studies have shown the association of CD44 with AF genesis. OBJECTIVE: The purpose of this study was to explore the potential role of CD44 and its related signaling in tachypacing-induced downregulation of LTCC. METHODS AND RESULTS: In vitro, tachypacing in atrium-derived myocytes (HL-1 cell line) induced activation (phosphorylation) of cyclic adenosine monophosphate response element-binding protein (CREB). Furthermore, tachypacing promoted an association between CREB and CD44 in HL-1 myocytes, which was documented in atrial tissues from patients with AF. Deletion and mutational analysis of the LTCC promoter along with chromatin immunoprecipitation revealed that cyclic adenosine monophosphate response element is essential for tachypacing-inhibited LTCC transcription. Tachypacing also hindered the binding of p-CREB to the promoter of LTCC. Blockade of CREB/CD44 signaling in HL-1 cells attenuated tachypacing-triggered downregulation of LTCC and shortening of APD. Atrial myocytes isolated from CD44-/- mice exhibited higher LTCC current and longer APD than did those from wild-type mice. Ex vivo, tachypacing caused less activation of CREB in CD44-/- mice than in wild-type mice. In vivo, burst atrial pacing stimulated less inducibility of AF in CREB inhibitor-treated mice than in controls. CONCLUSION: Tachypacing-induced CREB/CD44 signaling contributes to the suppression of LTCC, which provides valuable information about the pathogenesis of atrial modeling and AF.


Assuntos
Fibrilação Atrial/terapia , Remodelamento Atrial/fisiologia , Proteína de Ligação a CREB/genética , Canais de Cálcio Tipo L/genética , Estimulação Cardíaca Artificial/métodos , Regulação da Expressão Gênica , Receptores de Hialuronatos/genética , Animais , Fibrilação Atrial/genética , Fibrilação Atrial/metabolismo , Western Blotting , Proteína de Ligação a CREB/biossíntese , Canais de Cálcio Tipo L/biossíntese , Linhagem Celular , DNA/genética , Modelos Animais de Doenças , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Átrios do Coração/fisiopatologia , Receptores de Hialuronatos/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transdução de Sinais
2.
J Clin Invest ; 129(8): 3140-3152, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31162142

RESUMO

Elevated blood glucose (hyperglycemia) is a hallmark metabolic abnormality in diabetes. Hyperglycemia is associated with protein kinase A (PKA)-mediated stimulation of L-type Ca2+ channels in arterial myocytes resulting in increased vasoconstriction. However, the mechanisms by which glucose activates PKA remain unclear. Here, we showed that elevating extracellular glucose stimulates cAMP production in arterial myocytes, and that this was specifically dependent on adenylyl cyclase 5 (AC5) activity. Super-resolution imaging suggested nanometer proximity between subpopulations of AC5 and the L-type Ca2+ channel pore-forming subunit CaV1.2. In vitro, in silico, ex vivo and in vivo experiments revealed that this close association is critical for stimulation of L-type Ca2+ channels in arterial myocytes and increased myogenic tone upon acute hyperglycemia. This pathway supported the increase in L-type Ca2+ channel activity and myogenic tone in two animal models of diabetes. Our collective findings demonstrate a unique role for AC5 in PKA-dependent modulation of L-type Ca2+ channel activity and vascular reactivity during acute hyperglycemia and diabetes.


Assuntos
Adenilil Ciclases/metabolismo , Artérias Cerebrais/enzimologia , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/enzimologia , Hiperglicemia/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Adenilil Ciclases/genética , Animais , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/genética , Artérias Cerebrais/patologia , AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Hiperglicemia/genética , Hiperglicemia/patologia , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia
3.
J Neurosci ; 38(43): 9215-9227, 2018 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-30201773

RESUMO

Stac protein (named for its SH3- and cysteine-rich domains) was first identified in brain 20 years ago and is currently known to have three isoforms. Stac2, Stac1, and Stac3 transcripts are found at high, modest, and very low levels, respectively, in the cerebellum and forebrain, but their neuronal functions have been little investigated. Here, we tested the effects of Stac proteins on neuronal, high-voltage-activated Ca2+ channels. Overexpression of the three Stac isoforms eliminated Ca2+-dependent inactivation (CDI) of l-type current in rat neonatal hippocampal neurons (sex unknown), but not CDI of non-l-type current. Using heterologous expression in tsA201 cells (together with ß and α2-δ1 auxiliary subunits), we found that CDI for CaV1.2 and CaV1.3 (the predominant, neuronal l-type Ca2+ channels) was suppressed by all three Stac isoforms, whereas CDI for the P/Q channel, CaV2.1, was not. For CaV1.2, the inhibition of CDI by the Stac proteins appeared to involve their direct interaction with the channel's C terminus. Within the Stac proteins, a weakly conserved segment containing ∼100 residues and linking the structurally conserved PKC C1 and SH3_1 domains was sufficient to fully suppress CDI. The presence of CDI for l-type current in control neonatal neurons raised the possibility that endogenous Stac levels are low in these neurons and Western blotting indicated that the expression of Stac2 was substantially increased in adult forebrain and cerebellum compared with neonate. Together, our results indicate that one likely function of neuronal Stac proteins is to tune Ca2+ entry via neuronal l-type channels.SIGNIFICANCE STATEMENT Stac protein, first identified 20 years ago in brain, has recently been found to be essential for proper trafficking and function of the skeletal muscle l-type Ca2+ channel and is the site of mutations causing a severe, inherited human myopathy. In neurons, however, functions for Stac protein have remained unexplored. Here, we report that one likely function of neuronal Stac proteins is tuning Ca2+ entry via l-type, but not that via non-l-type, Ca2+ channels. Moreover, there is a large postnatal increase in protein levels of the major neuronal isoform (Stac2) in forebrain and cerebellum, which could provide developmental regulation of l-type channel Ca2+ signaling in these brain regions.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Sinalização do Cálcio/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo L/genética , Células Cultivadas , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Neurourol Urodyn ; 37(8): 2441-2451, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29806709

RESUMO

AIM: To determine the efficacy of human relaxin-2 (hRLX2) in reversing radiation-induced bladder fibrosis and lower urinary tract dysfunction (LUTD). Radiation cystitis is a consequence of radiotherapy for pelvic malignancies. Acutely, irradiation leads to reactive oxygen/nitrogen species in urothelial cells, apoptosis, barrier disruption, and inflammation. Chronically, this results in collagen deposition, bladder fibrosis, and attenuated storage and voiding functions. In severe cases, cystectomies are performed as current therapies do not reverse fibrosis. METHODS: We developed a mouse model for selective bladder irradiation (10 Gray; 1 Gy = 100 rads) resulting in chronic fibrosis within 6 weeks, with decreased bladder compliance, contractility, and overflow incontinence. Seven weeks post-irradiation, female C57Bl/6 mice were continuously infused with hRLX2 (400 µg/kg/day/14 days) or vehicle (saline) via subcutaneous osmotic pumps. Mice were evaluated in vivo using urine spot analysis, cystometrograms and external urethral sphincter electromyograms; and in vitro using length-tension measurements, Western blots, histology, and immunohistochemistry. RESULTS: hRLX2 reversed fibrosis, decreased collagen content, improved bladder wall architecture, and increased bladder compliance, detrusor smooth muscle Cav1.2 expression and detrusor contractility in mice with chronic radiation cystitis. hRLX2 treatment outcomes were likely caused by the activation of RXFP1/2 receptors which are expressed on the detrusor. CONCLUSION: hRLX2 may be a new therapeutic option for rescuing bladders with chronic radiation cystitis.


Assuntos
Cistite/tratamento farmacológico , Cistite/patologia , Relaxina/uso terapêutico , Doenças da Bexiga Urinária/tratamento farmacológico , Bexiga Urinária/patologia , Bexiga Urinária/efeitos da radiação , Animais , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/genética , Colágeno/metabolismo , Cistite/etiologia , Eletromiografia , Feminino , Fibrose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/fisiologia , Lesões por Radiação/complicações , Lesões por Radiação/tratamento farmacológico , Proteínas Recombinantes , Uretra/fisiopatologia , Bexiga Urinária/metabolismo , Doenças da Bexiga Urinária/etiologia , Incontinência Urinária/tratamento farmacológico , Incontinência Urinária/etiologia
5.
Heart Rhythm ; 15(5): 741-749, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29330129

RESUMO

BACKGROUND: In long QT syndrome type 2, women are more prone than men to the lethal arrhythmia torsades de pointes. We previously reported that 17ß-estradiol (E2) up-regulates L-type Ca2+ channels and current (ICa,L) (∼30%) in rabbit ventricular myocytes by a classic genomic mechanism mediated by estrogen receptor-α (ERα). In long QT syndrome type 2 (IKr blockade or bradycardia), the higher Ca2+ influx via ICa,L causes Ca2+ overload, spontaneous sarcoplasmic reticulum Ca2+ release, and reactivation of ICa,L that triggers early afterdepolarizations and torsades de pointes. OBJECTIVE: The purpose of this study was to investigate the molecular mechanisms whereby E2 up-regulates ICa,L, which are poorly understood. METHODS: H9C2 and rat myocytes were incubated with E2 ± ER antagonist, or inhibitors of downstream transcription factors, for 24 hours, followed by western blots of Cav1.2α1C and voltage-clamp measurements of ICa,L. RESULTS: Incubation of H9C2 cells with E2 (10-100 nM) increased ICa,L density and Cav1.2α1C expression, which were suppressed by the ER antagonist ICI182,780 (1 µM). Enhanced ICa,L and Cav1.2α1C expression by E2 was suppressed by inhibitors of phosphoinositide-3-kinase (Pi3K) (30 µM LY294002; P <.05) and Akt (5 µM MK2206) but not of mitogen-activated protein kinase (5 µM U0126) or protein kinase A (1 µM KT5720). E2 incubation increased p-CREB via the Pi3K/Akt pathway, reached a peak in 20 minutes (3-fold), and leveled off to 1.5-fold 24 hours later. Furthermore, a CREB decoy oligonucleotide inhibited E2-induced Cav1.2α1C expression, whereas membrane-impermeable E2 (E2-bovine serum albumin) was equally effective at Cav1.2α1C up-regulation as E2. CONCLUSION: Estradiol up-regulates Cav1.2α1C and ICa,L via plasma membrane ER and by activating Pi3K, Akt, and CREB signaling. The promoter regions of the CACNA1C gene (human-rabbit-rat) contain adjacent/overlapping binding sites for p-CREB and ERα, which suggests a synergistic regulation by these pathways.


Assuntos
Proteína de Ligação a CREB/genética , Canais de Cálcio Tipo L/genética , Estradiol/farmacologia , Regulação da Expressão Gênica , Síndrome do QT Longo/genética , Miócitos Cardíacos/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Western Blotting , Proteína de Ligação a CREB/biossíntese , Proteína de Ligação a CREB/efeitos dos fármacos , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/efeitos dos fármacos , Proteínas de Transporte/biossíntese , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Linhagem Celular , Cromonas/farmacologia , DNA/genética , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Estrogênios/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/patologia , Morfolinas/farmacologia , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
6.
Tumour Biol ; 39(5): 1010428317692244, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28459217

RESUMO

Resistance to conventional therapies and frequent recurrence are the major obstacles to the treatment of high-grade gliomas, including glioblastoma. Thus, the development of new therapeutic strategies to overcome these obstacles is necessary to improve the treatment outcomes. In this study, we found that verapamil, a pan-adenosine triphosphate-binding cassette transporter and L-type voltage-dependent calcium channel inhibitor, sensitized U87MG glioma cells to carmustine- and irradiation-induced senescence. Furthermore, our results indicated that verapamil treatment, in combination with carmustine and irradiation, rendered U87MG glioma cells and several patient-derived glioma stem cells more sensitive to therapy-induced senescence than individual or dual-combination treatments. When investigating the underlying mechanism, we found that verapamil treatment markedly decreased intracellular reactive oxygen species and calcium ion levels. Reactive oxygen species reduction with N-acetylcysteine, a reactive oxygen species scavenger, rendered U87MG glioma cells more sensitive to carmustine and irradiation whereas the protein kinase C agonist, phorbol 12-myristate 13-acetate, mitigated the effects of carmustine and irradiation. Taken together, our results indicate that verapamil may be a potent therapeutic sensitizer for increasing the effectiveness of glioblastoma treatment.


Assuntos
Carmustina/administração & dosagem , Glioma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Verapamil/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica , Cálcio/metabolismo , Canais de Cálcio Tipo L/biossíntese , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Terapia Combinada , Glioma/patologia , Glioma/radioterapia , Humanos , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/radioterapia , Espécies Reativas de Oxigênio/metabolismo
7.
Biol Trace Elem Res ; 174(2): 300-308, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27129314

RESUMO

A low concentration of boron (B) accelerates the proliferation and differentiation of mammalian osteoblasts. The aim of this study was to investigate the effects of 0.1 mM of B on the membrane function of osteoblastic cells in vitro. Genes involved in cell activity were investigated using gene expression microarray analyses. The Ca2+ influx and efflux were evaluated to demonstrate the activation of L-type Ca2+ channel for the Ca2+ influx, and that of Na+/K+-ATPase for the Ca2+ efflux. A real-time PCR analysis revealed that the messenger RNA (mRNA) expression of four mineralization-related genes was clearly increased after 3 days of culture with a B-supplemented culture medium. Using microarray analyses, five genes involved in cell proliferation and differentiation were upregulated compared to the control group. Regarding the Ca2+ influx, in the nifedipine-pretreated group, the relative fluorescence intensity for 1 min after adding B solution did not increase compared with that for 1 min before addition. In the control group, the relative fluorescence intensity was significantly increased compared with the experimental group (P < 0.05). Regarding the Ca2+ efflux, in the experimental group cultured in 0.1 mM of B-supplemented medium, the relative fluorescence intensity for 10 min after ouabain treatment revealed a significantly lower slope value compared with the control group (P < 0.01). This is the first study to demonstrate the acceleration of Ca2+ flux by B supplementation in osteoblastic cells. Cell membrane stability is related to the mechanism by which a very low concentration of B promotes the proliferation and differentiation of mammalian osteoblastic cells in vitro.


Assuntos
Boro/farmacologia , Cálcio/metabolismo , Osteoblastos/metabolismo , Canais de Cálcio Tipo L/biossíntese , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , ATPase Trocadora de Sódio-Potássio/biossíntese
8.
Tsitologiia ; 58(6): 444-50, 2016.
Artigo em Inglês, Russo | MEDLINE | ID: mdl-30192115

RESUMO

The experiments of modeling hypogravity using fluorescent microscopy have shown a decrease of expression of b1 of Na +/K+-ATPase and Ca2+-ATPase subunits and the increase in the insensitivity of synthesis of a1S subunit of the L-type Ca2+-channel of the plasmatic membrane, whereas the synthesis of a2 subunit of Na+/K+-ATPase does not change. In «fast¼ muscle only observed similar for «slow¼ muscle decrease in the expression of b1 subunit without changing other parameters were studied. However, the decrease in fluorescence b1 subunit due to spread of data was not statistically significant. Thus hypogravity adversely affects the functioning primarily skeletal muscles, providing static load.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Hipergravidade , Fibras Musculares de Contração Lenta/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , ATPase Trocadora de Sódio-Potássio/biossíntese , Animais , Masculino , Microscopia de Fluorescência , Fibras Musculares de Contração Lenta/patologia , Ratos , Ratos Wistar
9.
J Mol Cell Cardiol ; 86: 23-31, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26103619

RESUMO

In mammalian cardiac ventricular myocytes, Ca influx and release occur predominantly at t-tubules, ensuring synchronous Ca release throughout the cell. Heart failure is associated with disrupted t-tubule structure, but its effect on t-tubule function is less clear. We therefore investigated Ca influx and release at the t-tubules of ventricular myocytes isolated from rat hearts ~18weeks after coronary artery ligation (CAL) or corresponding Sham operation. L-type Ca current (ICa) was recorded using the whole-cell voltage-clamp technique in intact and detubulated myocytes; Ca release at t-tubules was monitored using confocal microscopy with voltage- and Ca-sensitive fluorophores. CAL was associated with cardiac and cellular hypertrophy, decreased ejection fraction, disruption of t-tubule structure and a smaller, slower Ca transient, but no change in ryanodine receptor distribution, L-type Ca channel expression, or ICa density. In Sham myocytes, ICa was located predominantly at the t-tubules, while in CAL myocytes, it was uniformly distributed between the t-tubule and surface membranes. Inhibition of protein kinase A with H-89 caused a greater decrease of t-tubular ICa in CAL than in Sham myocytes; in the presence of H-89, t-tubular ICa density was smaller in CAL than in Sham myocytes. The smaller t-tubular ICa in CAL myocytes was accompanied by increased latency and heterogeneity of SR Ca release at t-tubules, which could be mimicked by decreasing ICa using nifedipine. These data show that CAL decreases t-tubular ICa via a PKA-independent mechanism, thereby impairing Ca release at t-tubules and contributing to the altered excitation-contraction coupling observed in heart failure.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Cálcio/metabolismo , Insuficiência Cardíaca/metabolismo , Ventrículos do Coração/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Insuficiência Cardíaca/patologia , Ventrículos do Coração/patologia , Humanos , Isoquinolinas/administração & dosagem , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Ratos , Sulfonamidas/administração & dosagem
10.
J Neurochem ; 134(1): 56-65, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25807982

RESUMO

In neurons, calcium (Ca(2+) ) channels regulate a wide variety of functions ranging from synaptic transmission to gene expression. They also induce neuroplastic changes that alter gene expression following psychostimulant administration. Ca(2+) channel blockers have been considered as potential therapeutic agents for the treatment of methamphetamine (METH) dependence because of their ability to reduce drug craving among METH users. Here, we studied the effects of METH exposure on voltage-gated Ca(2+) channels using SH-SY5Y cells as a model of dopaminergic neurons. We found that METH has different short- and long-term effects. A short-term effect involves immediate (< 5 min) direct inhibition of Ca(2+) ion movements through Ca(2+) channels. Longer exposure to METH (20 min or 48 h) selectively up-regulates the expression of only the CACNA1C gene, thus increasing the number of L-type Ca(2+) channels. This up-regulation of CACNA1C is associated with the expression of the cAMP-responsive element-binding protein (CREB), a known regulator of CACNA1C gene expression, and the MYC gene, which encodes a transcription factor that putatively binds to a site proximal to the CACNA1C gene transcription initiation site. The short-term inhibition of Ca(2+) ion movement and later, the up-regulation of Ca(2+) channel gene expression together suggest the operation of cAMP-responsive element-binding protein- and C-MYC-mediated mechanisms to compensate for Ca(2+) channel inhibition by METH. Increased Ca(2+) current density and subsequent increased intracellular Ca(2+) may contribute to the neurodegeneration accompanying chronic METH abuse. Methamphetamine (METH) exposure has both short- and long-term effects. Acutely, methamphetamine directly inhibits voltage-gated calcium channels. Chronically, neurons compensate by up-regulating the L-type Ca(2+) channel gene, CACNA1C. This compensatory mechanism is mediated by transcription factors C-MYC and CREB, in which CREB is linked to the dopamine D1 receptor signaling pathway. These findings suggest Ca(2+) -mediated neurotoxicity owing to over-expression of calcium channels.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/biossíntese , Metanfetamina/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Linhagem Celular Tumoral , Humanos , Fatores de Tempo
11.
Stem Cell Rev Rep ; 11(3): 408-22, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25277766

RESUMO

Bone marrow-derived mesenchymal stem cells (MSC) can be differentiated into myocytes, as well as adipocytes, chondrocytes, and osteocytes in culture. Calcium channels mediate excitation-contraction coupling and are essential for the function of muscle. However, little is known about the expression of calcium channel subunits and calcium handling in stem cells. We examined whether the expression of calcium channel subunits in MSC is similar to that of skeletal muscle satellite cells and if their levels of expression are modified after treatment with bone morphogenetic protein-4 (BMP4). We found that during myogenic differentiation, MSC first express the α2δ1 subunit and the cardiac channel subunit Cav1.2. In contrast to the α2δ1 subunit levels, the Cav1.2 subunit decreases rapidly with time. The skeletal channel subunit Cav1.1 is detected at day 3 but its expression increases considerably, resembling more closely the expression of the subunits in satellite cells. Treatment of MSC with BMP4 caused a significant increase in expression of Cav1.2, a delay in expression of Cav1.1, and a reduction in the duration of calcium transients when extracellular calcium was removed. Calcium currents and transients followed a pattern related to the expression of the cardiac (Cav1.2) or skeletal (Cav1.1) α1subunits. These results indicate that differentiation of untreated MSC resembles differentiation of skeletal muscle and that BMP4 reduces skeletal muscle calcium channel expression and promotes the expression of cardiac calcium channels during myogenic differentiation.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Canais de Cálcio Tipo L/biossíntese , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 4/administração & dosagem , Canais de Cálcio Tipo L/genética , Sinalização do Cálcio , Condrócitos/citologia , Condrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Camundongos , Desenvolvimento Muscular/genética , Músculo Esquelético/citologia , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo
12.
J Cell Physiol ; 229(12): 1926-34, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24700505

RESUMO

Primary cilia are sensory organelles that provide a feedback mechanism to restrict Wnt signaling in the absence of endogenous Wnt activators. Abnormal Wnt signaling has been shown to result in polycystic kidney disease (PKD) although the exact mechanism has been debated. Previously, we reported that the calcium channel CaV1.2 functions in primary cilia. In this study, we show that CaV1.2 expression level is regulated by Wnt signaling. This occurs through modulation of mitochondrial mass and activity resulting in increased reactive oxygen species which generate oxidative DNA lesions. We found that the subsequent cellular DNA damage response triggers increased CaV1.2 expression. In the absence of primary cilia where Wnt signaling is upregulated, we found that CaV1.2 is overexpressed as a compensatory mechanism. We show for the first time that CaV1.2 knockdown in zebrafish results in classic primary cilia defects including renal cyst formation, hydrocephalus, and left-right asymmetry defects. Our study shows that suppressed Wnt signaling prevents CaV1.2 expression ultimately resulting in PKD phenotypes. Thus, CaV1.2 expression is tightly regulated through Wnt signaling and plays an essential sensory role in primary cilia necessary for cellular homeostasis.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Cílios/metabolismo , Doenças Renais Policísticas/metabolismo , Via de Sinalização Wnt/genética , Animais , Canais de Cálcio Tipo L/genética , Cílios/genética , Regulação da Expressão Gênica , Humanos , Rim/metabolismo , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/patologia , Espécies Reativas de Oxigênio/metabolismo , Peixe-Zebra
13.
J Cardiothorac Surg ; 8: 194, 2013 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-24074263

RESUMO

BACKGROUND: Recent studies have demonstrated that atrial electrical remodeling was an important contributing factor for the occurrence, persistence and maintenance of atrial fibrillation. The expression changes of ionic channels, especially L-type calcium channel and potassium channel Kv4.3, were the important molecular mechanism of atrial electrical remodeling. This study aimed to observe the expression changes of ionic channels in a rapid paced cell model with primary cultured atrial myocytes. METHODS: The primary rat atrial myocytes were cultured, characteristics of the cultured myocytes were observed with light microscope and the cell phenotype was harvested by immunocytochemical stain to detect α-actin. The cellular model of rapid pacing was established with primary cultured atrial myocytes. The expressions of L-type calcium channel α1c and potassium channel Kv4.3 in cultured atrial myocytes were detected by immunocytochemistry, reverse transcription polymerase chain reaction and Western blot after rapid pacing. RESULTS: The primary rat atrial myocytes were isolated and cultured successfully, and used for following experiment by identification of activity and purity. Cellular model of rapid electrical field pacing was established successfully. There is no significant difference in cell activity after pacing compared to that before pacing by 3-[4, 5-dimethylthiazol-2-y1]-2, 5-diphenytetrazolium bromide assay, and cell degeneration can be observed by transmission electron microscope. The mRNA expression of L-type calcium channel α1c started to reduce after 6 h of rapid pacing and continued to decline as pacing continued. Protein expression changes were paralleled with decreased mRNA expression of the L-type calcium channel α1c. The mRNA expressions of potassium channel Kv4.3 were not altered within the first 6 h, but after 12 h, mRNA expressions were reduced. Longer pacing periods did not further decrease mRNA expression of potassium channel Kv4.3. Protein expression changes were paralleled with decreased mRNA expression of potassium channel Kv4.3. CONCLUSIONS: Rapid paced cultured atrial myocyte model was established utilized primary cultured atrial myocytes and this model can be used for studying the early electrical remodeling in atrial fibrillation. Expressions of L-type calcium channel α1c and potassium channel Kv4.3 were both reduced at different levels in early phase of rapid pacing atrial myocytes. It implicates the occurrence of ionic channel remodeling of atrial myocytes.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Estimulação Cardíaca Artificial/métodos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Canais de Potássio Shal/biossíntese , Actinas/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Células Cultivadas , Estimulação Elétrica , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Imuno-Histoquímica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Canais de Potássio Shal/genética
14.
Int J Nanomedicine ; 8: 93-107, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23319861

RESUMO

INTRODUCTION: In this study, quasi-three-dimensional (3D) microwell patterns were fabricated with poly (l-lactic acid) for the development of cell-based assays, targeting voltage-gated calcium channels (VGCCs). METHODS AND MATERIALS: SH-SY5Y human neuroblastoma cells were interfaced with the microwell patterns and found to grow as two dimensional (2D), 3D, and near two dimensional (N2D), categorized on the basis of the cells' location in the pattern. The capability of the microwell patterns to support 3D cell growth was evaluated in terms of the percentage of the cells in each growth category. Cell spreading was analyzed in terms of projection areas under light microscopy. SH-SY5Y cells' VGCC responsiveness was evaluated with confocal microscopy and a calcium fluorescent indicator, Calcium Green™-1. The expression of L-type calcium channels was evaluated using immunofluorescence staining with DM-BODIPY. RESULTS: It was found that cells within the microwells, either N2D or 3D, showed more rounded shapes and less projection areas than 2D cells on flat poly (l-lactic acid) substrates. Also, cells in microwells showed a significantly lower VGCC responsiveness than cells on flat substrates, in terms of both response magnitudes and percentages of responsive cells, upon depolarization with 50 mM K(+). This lower VGCC responsiveness could not be explained by the difference in L-type calcium channel expression. For the two patterns addressed in this study, N2D cells consistently exhibited an intermediate value of either projection areas or VGCC responsiveness between those for 2D and 3D cells, suggesting a correlative relation between cell morphology and VGCC responsiveness. CONCLUSION: These results suggest that the pattern structure and therefore the cell growth characteristics were critical factors in determining cell VGCC responsiveness and thus provide an approach for engineering cell functionality in cell-based assay systems and tissue engineering scaffolds.


Assuntos
Bioengenharia/métodos , Canais de Cálcio Tipo L/metabolismo , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Ácido Láctico/química , Neuroblastoma/metabolismo , Polímeros/química , Análise de Variância , Bioengenharia/instrumentação , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/química , Linhagem Celular Tumoral , Humanos , Microscopia Confocal , Neuroblastoma/patologia , Compostos Orgânicos/química , Poliésteres , Estatísticas não Paramétricas
15.
Dev Neurosci ; 34(5): 440-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23257417

RESUMO

The voltage-gated calcium channel subunit α(2)δ plays a fundamental role in propagation of excitatory signals associated with release of glutamate and neuropeptides substance P (SP) and calcitonin gene-related protein (CGRP). It can be selectively inhibited by gabapentinoids. Hence, investigation of the α(2)δ subunit may predict the efficacy of gabapentinoid therapy in neuropathic pain. Since sensory processing underlies significant age-related changes, this study was conducted in order to elucidate the role of the α(2)δ subunit in the sensory transmission during canine development. Dorsal root ganglia (DRG) were harvested from four spinal segments of 16 puppies and 10 adult dogs without a history of neurological signs, pain, spinal disease or orthopedic disorders. α(2)δ-Subunit expression and coexpression with SP and CGRP was evaluated immunohistochemically regarding the number of immunopositive ganglion cells, staining intensity and subcellular distribution. All tested ganglia were immunopositive for α(2)δ. Cell counts and expression levels were significantly lower in pups than in adult dogs (p < 0.05). In the cervical segments of both groups, the number and percentage of immunopositive neurons was significantly higher than in lumbar DRG (p < 0.05). Multilabeling studies in all tested animals confirmed the coexpression of α(2)δ and pain peptides SP and CGRP. This anatomical study for the first time documents the involvement of α(2)δ subunits in sensory signal processing in dogs. The proportion of positive neurons and the intracellular expression levels show a net increase from early postnatal life to adulthood. A significant portion of α(2)δ-positive cells in the dogs exhibited C- and Aδ-phenotypes compatible with nociceptive neurons. The coexpression of α(2)δ, SP and CGRP imply that these neurons are involved with peptidergic nociception. The cervicolumbar gradient of α(2)δ expression in adults reflects functional differences in between forelimbs and hind limbs. These data will facilitate translational studies on neuropathic pain states in this species such as common canine nerve entrapment syndromes.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/metabolismo , Animais , Biomarcadores , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Canais de Cálcio Tipo L/genética , Tamanho Celular , Doenças do Cão/tratamento farmacológico , Doenças do Cão/genética , Cães , Membro Anterior/crescimento & desenvolvimento , Membro Anterior/inervação , Gânglios Espinais/citologia , Membro Posterior/crescimento & desenvolvimento , Membro Posterior/inervação , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Neuralgia/tratamento farmacológico , Neuralgia/genética , Neuralgia/veterinária , Neurônios/ultraestrutura , Medula Espinal/citologia , Substância P/metabolismo
16.
J Physiol Sci ; 62(5): 385-92, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22695819

RESUMO

Our previous work has shown that interleukin-6 (IL-6) implements its neuroprotective effect by inhibiting the intracellular Ca(2+) overload in neurons. Here, we examined whether regulation of L-type calcium channels (LCCs) activities is involved in the neuroprotective action of IL-6. In cultured cerebellar granule neurons (CGNs), patch-clamp recording showed that the whole-cell Ca(2+) current and LCC current were significantly reduced by IL-6 pretreatment (120 ng/ml, for 24 h). Calcium imaging data indicated that IL-6 significantly suppressed high K(+)-induced intracellular Ca(2+) overload and LCC Ca(2+) influx. Moreover, expression of the LCC subunit, Ca(v)1.2, was remarkably downregulated by IL-6 in cultured CGNs. These findings suggest that IL-6 exerts a neurotrophic effect by preventing Ca(2+) overload, at least partly through inhibition of LCC activity in cultured CGNs.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Cerebelo/efeitos dos fármacos , Citoproteção , Interleucina-6/farmacologia , Neurônios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Células Cultivadas , Cerebelo/citologia , Neurônios/citologia , Técnicas de Patch-Clamp , Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia
17.
J Comp Neurol ; 520(16): 3650-72, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22473424

RESUMO

Precise axon pathfinding is crucial for establishment of the initial neuronal network during development. Pioneer axons navigate without the help of preexisting axons and pave the way for follower axons that project later. Voltage-gated ion channels make up the intrinsic electrical activity of pioneer axons and regulate axon pathfinding. To elucidate which channel molecules are present in pioneer axons, immunohistochemical analysis was performed to examine 14 voltage-gated ion channels (Kv1.1-Kv1.3, Kv3.1-Kv3.4, Kv4.3, Cav1.2, Cav1.3, Cav2.2, Nav1.2, Nav1.6, and Nav1.9) in nine axonal tracts in the developing rat forebrain, including the optic nerve, corpus callosum, corticofugal fibers, thalamocortical axons, lateral olfactory tract, hippocamposeptal projection, anterior commissure, hippocampal commissure, and medial longitudinal fasciculus. We found A-type K⁺ channel Kv3.4 in both pioneer axons and early follower axons and L-type Ca²âº channel Cav1.2 in pioneer axons and early and late follower axons. Spatially, Kv3.4 and Cav1.2 were colocalized with markers of pioneer neurons and pioneer axons, such as deleted in colorectal cancer (DCC), in most fiber tracts examined. Temporally, Kv3.4 and Cav1.2 were expressed abundantly in most fiber tracts during axon pathfinding but were downregulated beginning in synaptogenesis. By contrast, delayed rectifier Kv channels (e.g., Kv1.1) and Nav channels (e.g., Nav1.2) were absent from these fiber tracts (except for the corpus callosum) during pathfinding of pioneer axons. These data suggest that Kv3.4 and Cav1.2, two high-voltage-activated ion channels, may act together to control Ca²âº -dependent electrical activity of pioneer axons and play important roles during axon pathfinding.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Prosencéfalo/embriologia , Canais de Potássio Shaw/biossíntese , Animais , Movimento Celular , Feminino , Imunofluorescência , Imuno-Histoquímica , Masculino , Microscopia Confocal , Neurogênese/fisiologia , Prosencéfalo/metabolismo , Ratos , Ratos Sprague-Dawley
18.
J Biol Chem ; 286(18): 15757-65, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21402706

RESUMO

Insulin release by pancreatic ß-cells is regulated by diverse intracellular signals, including changes in Ca(2+) concentration resulting from Ca(2+) entry through voltage-gated (Ca(V)) channels. It has been reported that the Rab3 effector RIM1 acts as a functional link between neuronal Ca(V) channels and the machinery for exocytosis. Here, we investigated whether RIM1 regulates recombinant and native L-type Ca(V) channels (that play a key role in hormone secretion) and whether this regulation affects insulin release. Whole-cell patch clamp currents were recorded from HEK-293 and insulinoma RIN-m5F cells. RIM1 and Ca(V) channel expression was identified by RT-PCR and Western blot. RIM1-Ca(V) channel interaction was determined by co-immunoprecipitation. Knockdown of RIM1 and Ca(V) channel subunit expression were performed using small interference RNAs. Insulin release was assessed by ELISA. Co-expression of Ca(V)1.2 and Ca(V)1.3 L-type channels with RIM1 in HEK-293 cells revealed that RIM1 may not determine the availability of L-type Ca(V) channels but decreases the rate of inactivation of the whole cell currents. Co-immunoprecipitation experiments showed association of the Ca(V)ß auxiliary subunit with RIM1. The lack of Ca(V)ß expression suppressed channel regulation by RIM1. Similar to the heterologous system, an increase of current inactivation was observed upon knockdown of endogenous RIM1. Co-immunoprecipitation showed association of Ca(V)ß and RIM1 in insulin-secreting RIN-m5F cells. Knockdown of RIM1 notably impaired high K(+)-stimulated insulin secretion in the RIN-m5F cells. These data unveil a novel functional coupling between RIM1 and the L-type Ca(V) channels via the Ca(V)ß auxiliary subunit that contribute to determine insulin secretion.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canais de Cálcio Tipo L/genética , Linhagem Celular Tumoral , Proteínas de Ligação ao GTP/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Insulina/genética , Secreção de Insulina
19.
J Biol Chem ; 286(12): 10007-16, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21282112

RESUMO

CaV1.2 calcium channels play roles in diverse cellular processes such as gene regulation, muscle contraction, and membrane excitation and are diversified in their activity through extensive alternative splicing of the CaV1.2 mRNA. The mutually exclusive exons 8a and 8 encode alternate forms of transmembrane segment 6 (IS6) in channel domain 1. The human genetic disorder Timothy syndrome is caused by mutations in either of these two CaV1.2 exons, resulting in disrupted Ca(2+) homeostasis and severe pleiotropic disease phenotypes. The tissue-specific pattern of exon 8/8a splicing leads to differences in symptoms between patients with exon 8 or 8a mutations. Elucidating the mechanisms controlling the exon 8/8a splicing choice will be important in understanding the spectrum of defects associated with the disease. We found that the polypyrimidine tract-binding protein (PTB) mediates a switch from exon 8 to 8a splicing. PTB and its neuronal homolog, nPTB, are widely studied splicing regulators controlling large sets of alternative exons. During neuronal development, PTB expression is down-regulated with a concurrent increase in nPTB expression. Exon 8a is largely repressed in embryonic mouse brain but is progressively induced during neuronal differentiation as PTB is depleted. This splicing repression is mediated by the direct binding of PTB to sequence elements upstream of exon 8a. The nPTB protein is a weaker repressor of exon 8a, resulting in a shift in exon choice when nPTB replaces PTB in cells. These results provide mechanistic understanding of how these two exons, important for human disease, are controlled.


Assuntos
Processamento Alternativo/fisiologia , Canais de Cálcio Tipo L/biossíntese , Éxons/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Animais , Transtorno Autístico , Encéfalo/embriologia , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Estrutura Terciária de Proteína , Sindactilia/genética , Sindactilia/metabolismo
20.
J Mol Cell Cardiol ; 50(3): 460-70, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21111744

RESUMO

Pathological cardiac hypertrophy (PCH) is associated with the development of arrhythmia and congestive heart failure. While calcium (Ca(2+)) is implicated in hypertrophic signaling pathways, the specific role of Ca(2+) influx through the L-type Ca(2+) channel (I(Ca-L)) has been controversial and is the topic of this study. To determine if and how sustained increases in I(Ca-L) induce PCH, transgenic mouse models with low (LE) and high (HE) expression levels of the ß2a subunit of Ca(2+) channels (ß2a) and in cultured adult feline (AF) and neonatal rat (NR) ventricular myocytes (VMs) infected with an adenovirus containing a ß2a-GFP were used. In vivo, ß2a LE and HE mice had increased heart weight to body weight ratio, posterior wall and interventricular septal thickness, tissue fibrosis, myocyte volume, and cross-sectional area and the expression of PCH markers in a time- and dose-dependent manner. PCH was associated with a hypercontractile phenotype including enhanced I(Ca-L), fractional shortening, peak Ca(2+) transient, at the myocyte level, greater ejection fraction, and fractional shortening at the organ level. In addition, LE mice had an exaggerated hypertrophic response to transverse aortic constriction. In vitro overexpression of ß2a in cultured AFVMs increased I(Ca-L), cell volume, protein synthesis, NFAT, and HDAC translocations and in NRVMs increased surface area. These effects were abolished by the blockade of I(Ca-L), intracellular Ca(2+), calcineurin, CaMKII, and SERCA. In conclusion, increasing I(Ca-L) is sufficient to induce PCH through the calcineurin/NFAT and CaMKII/HDAC pathways. Both cytosolic and SR/ER-nuclear envelop Ca(2+) pools were shown to be involved.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Animais , Calcineurina/metabolismo , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Gatos , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Histona Desacetilases/metabolismo , Camundongos , Camundongos Transgênicos , Contração Miocárdica , Fatores de Transcrição NFATC/metabolismo , Membrana Nuclear/metabolismo , Fenótipo , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA