Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 610
Filtrar
Más filtros

Publication year range
1.
J Physiol ; 601(14): 2935-2958, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37278367

RESUMEN

The acrosome is a lysosome-related vesicular organelle located in the sperm head. The acrosomal reaction (AR) is an exocytic process mediated by Ca2+ and essential for mammalian fertilization. Recent findings support the importance of acrosomal alkalinization for the AR. Mibefradil (Mib) and NNC 55-0396 (NNC) are two amphipathic weak bases that block the sperm-specific Ca2+ channel (CatSper) and induce acrosomal pH (pHa ) increase by accumulating in the acrosomal lumen of mammalian sperm. This accumulation and pHa elevation increase the intracellular Ca2+ concentration ([Ca2+ ]i ) and trigger the AR by unknown mechanisms of Ca2+ transport. Here, we investigated the pathways associated with the pHa increase-induced Ca2+ signals using mouse sperm as a model. To address these questions, we used single-cell Ca2+ imaging, the lysosomotropic agent Gly-Phe-ß-naphthylamide (GPN) and pharmacological tools. Our findings show that Mib and NNC increase pHa and release acrosomal Ca2+ without compromising acrosomal membrane integrity. Our GPN results indicate that the osmotic component does not significantly contribute to acrosomal Ca2+ release caused by pHa rise. Inhibition of two-pore channel 1 (TPC1) channels reduced the [Ca2+ ]i increase stimulated by acrosomal alkalinization. In addition, blockage of Ca2+ release-activated Ca2+ (CRAC) channels diminished Ca2+ uptake triggered by pHa alkalinization. Finally, our findings contribute to understanding how pHa controls acrosomal Ca2+ efflux and extracellular Ca2+ entry during AR in mouse sperm. KEY POINTS: The acrosomal vesicle is a lysosome-related organelle located in the sperm head. The acrosome reaction (AR) is a highly regulated exocytic process mediated by Ca2+ , which is essential for fertilization. However, the molecular identity of Ca2+ transporters involved in the AR and their mechanisms to regulate Ca2+ fluxes are not fully understood. In mammalian sperm, acrosomal alkalinization induces intracellular Ca2+ concentration ([Ca2+ ]i ) increase and triggers the AR by unknown molecular mechanisms of Ca2+ transport. In this study, we explored the molecular mechanisms underlying Ca2+ signals caused by acrosomal alkalinization using mouse sperm as a model. TPC1 and CRAC channels contribute to [Ca2+ ]i elevation during acrosomal alkalinization. Our findings expand our understanding of how the acrosomal pH participates in the physiological induction of the AR.


Asunto(s)
Calcio , Semen , Masculino , Animales , Ratones , Calcio/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo , Acrosoma/metabolismo , Mibefradil/metabolismo , Mibefradil/farmacología , Concentración de Iones de Hidrógeno , Mamíferos/metabolismo
2.
Int Arch Allergy Immunol ; 183(6): 579-590, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35100604

RESUMEN

INTRODUCTION: The mucociliary transport function of the airway epithelium is largely dependent on ciliary beating. The control signal of ciliary beating is thought to be intracellular Ca2+. We herein investigated the expression of T-type voltage-gated calcium channel (VGCC), a generator of intracellular Ca2+ oscillation, in the human nasal mucosa. METHODS: The inferior turbinate was collected from patients with chronic hypertrophic rhinitis. The expression of T-type VGCC α1 subunits was examined by immunohistochemistry, transmission immunoelectron microscopy, Western blot, and real-time reverse transcription-polymerase chain reaction (RT-PCR). Participation of T-type VGCC in the ciliary beat regulation was examined by pharmacological inhibition tests using specific blockers of T-type VGCC in ex vivo measurements of the ciliary beat frequency (CBF) and ATP release and in intracellular Ca2+ imaging of isolated ciliated cells. RESULTS: Immunohistochemical staining showed the expressions of T-type VGCC α1 subunits, Cav3.1 and Cav3.3, on the surface of the epithelial cells. At the ultrastructural level, immunoreactivity for Cav3.1 was localized on the surface of the cilia, and that for Cav3.3 was localized in the cilia and at the base of the cilia. The existence of Cav3.1 and Cav3.3 was confirmed at the protein level by Western blot and at the transcriptional level by real-time RT-PCR. Specific blockers of T-type VGCC, mibefradil and NNC 55-0396, significantly inhibited CBF. These blockers also inhibited a CBF increase induced by 8-bromo-cAMP/8-bromo-cGMP and significantly lowered the intracellular Ca2+ level of isolated ciliated cells in a time-dependent manner. On the other hand, the ATP release from the nasal mucosa was not changed by mibefradil or NNC 55-0396. CONCLUSION: These results indicate that T-type VGCC α1 subunits, Cav3.1 and Cav3.3, exist at the cilia of the nasal epithelial cells and participate in the regulation of ciliary beating and that these channels act downstream of cAMP/cGMP.


Asunto(s)
Canales de Calcio Tipo T , Cilios , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Cilios/fisiología , GMP Cíclico , Células Epiteliales/metabolismo , Humanos , Mibefradil/metabolismo , Mibefradil/farmacología , Mucosa Nasal/metabolismo
3.
Clin Exp Pharmacol Physiol ; 49(1): 25-34, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34438468

RESUMEN

Atrial fibrillation (AF) is associated with atrial conduction disturbances caused by electrical and/or structural remodelling. In the present study, we hypothesized that connexin might interact with the calcium channel through forming a protein complex and, then, participates in the pathogenesis of AF. Western blot and whole-cell patch clamp showed that protein levels of Cav1.2 and connexin 43 (Cx43) and basal ICa,L were decreased in AF subjects compared to sinus rhythm (SR) controls. In cultured atrium-derived myocytes (HL-1 cells), knocking-down of Cx43 or incubation with 30 mmol/L glycyrrhetinic acid significantly inhibited protein levels of Cav1.2 and Cav3.1 and the current density of ICa,L and ICa,T . Incubation with nifedipine or mibefradil decreased the protein level of Cx43 in HL-1 cells. Moreover, Cx43 was colocalized with Cav1.2 and Cav3.1 in atrial myocytes. Therefore, Cx43 might regulate the ICa,L and ICa,T through colocalization with calcium channel subunits in atrial myocytes, representing a potential pathogenic mechanism in AF.


Asunto(s)
Remodelación Atrial , Canales de Calcio/fisiología , Conexina 43/fisiología , Atrios Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Fibrilación Atrial/metabolismo , Remodelación Atrial/fisiología , Western Blotting , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/fisiología , Línea Celular , Células Cultivadas , Conexina 43/metabolismo , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/fisiopatología , Humanos , Mibefradil/farmacología , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Nifedipino/farmacología , Técnicas de Placa-Clamp
4.
BMC Nephrol ; 23(1): 211, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35710406

RESUMEN

BACKGROUND: T-type calcium channels (TTCC) are low voltage activated channels that are widely expressed in the heart, smooth muscle and neurons. They are known to impact on cell cycle progression in cancer and smooth muscle cells and more recently, have been implicated in rat and human mesangial cell proliferation. The aim of this study was to investigate the roles of the different isoforms of TTCC in mouse mesangial cells to establish which may be the best therapeutic target for treating mesangioproliferative kidney diseases.  METHODS: In this study, we generated single and double knockout (SKO and DKO) clones of the TTCC isoforms CaV3.1 and CaV3.2 in mouse mesangial cells using CRISPR-cas9 gene editing. The downstream signals linked to this channel activity were studied by ERK1/2 phosphorylation assays in serum, PDGF and TGF-ß1 stimulated cells. We also examined their proliferative responses in the presence of the TTCC inhibitors mibefradil and TH1177. RESULTS: We demonstrate a complete loss of ERK1/2 phosphorylation in response to multiple stimuli (serum, PDGF, TGF-ß1) in CaV3.1 SKO clone, whereas the CaV3.2 SKO clone retained these phospho-ERK1/2 responses. Stimulated cell proliferation was not profoundly impacted in either SKO clone and both clones remained sensitive to non-selective TTCC blockers, suggesting a role for more than one TTCC isoform in cell cycle progression. Deletion of both the isoforms resulted in cell death. CONCLUSION: This study confirms that TTCC are expressed in mouse mesangial cells and that they play a role in cell proliferation. Whereas the CaV3.1 isoform is required for stimulated phosphorylation of ERK1/2, the Ca V3.2 isoform is not. Our data also suggest that neither isoform is necessary for cell proliferation and that the anti-proliferative effects of mibefradil and TH1177 are not isoform-specific. These findings are consistent with data from in vivo rat mesangial proliferation Thy1 models and support the future use of genetic mouse models to test the therapeutic actions of TTCC inhibitors.


Asunto(s)
Canales de Calcio Tipo T , Células Mesangiales , Animales , Humanos , Células Mesangiales/metabolismo , Mibefradil/metabolismo , Mibefradil/farmacología , Ratones , Fosforilación , Ratas , Factor de Crecimiento Transformador beta1/metabolismo
5.
Int J Mol Sci ; 23(17)2022 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-36077291

RESUMEN

The role of T-type calcium channels is well established in excitable cells, where they preside over action potential generation, automaticity, and firing. They also contribute to intracellular calcium signaling, cell cycle progression, and cell fate; and, in this sense, they emerge as key regulators also in non-excitable cells. In particular, their expression may be considered a prognostic factor in cancer. Almost all cancer cells express T-type calcium channels to the point that it has been considered a pharmacological target; but, as the drugs used to reduce their expression are not completely selective, several complications develop, especially within the heart. T-type calcium channels are also involved in a specific side effect of several anticancer agents, that act on microtubule transport, increase the expression of the channel, and, thus, the excitability of sensory neurons, and make the patient more sensitive to pain. This review puts into context the relevance of T-type calcium channels in cancer and in chemotherapy side effects, considering also the cardiotoxicity induced by new classes of antineoplastic molecules.


Asunto(s)
Canales de Calcio Tipo T , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo T/metabolismo , Señalización del Calcio , Humanos , Mibefradil/farmacología
6.
Int J Mol Sci ; 23(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36555708

RESUMEN

Notch3 plays an important role in the differentiation and development of vascular smooth muscle cells. Mice lacking Notch3 show deficient renal autoregulation. The aim of the study was to investigate the mechanisms involved in the Notch3-mediated control of renal vascular response. To this end, renal resistance vessels (afferent arterioles) were isolated from Notch3-/- and wild-type littermates (WT) and stimulated with angiotensin II (ANG II). Contractions and intracellular Ca2+ concentrations were blunted in Notch3-/- vessels. ANG II responses in precapillary muscle arterioles were similar between the WT and Notch3-/- mice, suggesting a focal action of Notch3 in renal vasculature. Abolishing stored Ca2+ with thapsigargin reduced Ca2+ responses in the renal vessels of the two strains, signifying intact intracellular Ca2+ mobilization in Notch3-/-. EGTA (Ca2+ chelating agent), nifedipine (L-type channel-blocker), or mibefradil (T-type channel-blocker) strongly reduced contraction and Ca2+ responses in WT mice but had no effect in Notch3-/- mice, indicating defective Ca2+ entry. Notch3-/- vessels responded normally to KCl-induced depolarization, which activates L-type channels directly. Differential transcriptomic analysis showed a major down-regulation of Cacna1h gene expression, coding for the α1H subunit of the T-type Ca2+ channel, in Notch3-/- vessels. In conclusion, renal resistance vessels from Notch3-/- mice display altered vascular reactivity to ANG II due to deficient Ca2+-entry. Consequently, Notch3 is essential for proper excitation-contraction coupling and vascular-tone regulation in the kidney.


Asunto(s)
Riñón , Nifedipino , Receptor Notch3 , Animales , Ratones , Angiotensina II/farmacología , Arteriolas/metabolismo , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Riñón/metabolismo , Mibefradil/metabolismo , Nifedipino/farmacología , Resistencia Vascular , Receptor Notch3/genética , Eliminación de Gen , Ratones Noqueados
7.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-33924361

RESUMEN

TRPM7 plays an important role in cellular Ca2+, Zn2+ and Mg2+ homeostasis. TRPM7 channels are abundantly expressed in ameloblasts and, in the absence of TRPM7, dental enamel is hypomineralized. The potential role of TRPM7 channels in Ca2+ transport during amelogenesis was investigated in the HAT-7 rat ameloblast cell line. The cells showed strong TRPM7 mRNA and protein expression. Characteristic TRPM7 transmembrane currents were observed, which increased in the absence of intracellular Mg2+ ([Mg2+]i), were reduced by elevated [Mg2+]i, and were inhibited by the TRPM7 inhibitors NS8593 and FTY720. Mibefradil evoked similar currents, which were suppressed by elevated [Mg2+]i, reducing extracellular pH stimulated transmembrane currents, which were inhibited by FTY720. Naltriben and mibefradil both evoked Ca2+ influx, which was further enhanced by the acidic intracellular conditions. The SOCE inhibitor BTP2 blocked Ca2+ entry induced by naltriben but not by mibefradil. Thus, in HAT-7 cells, TRPM7 may serves both as a potential modulator of Orai-dependent Ca2+ uptake and as an independent Ca2+ entry pathway sensitive to pH. Therefore, TRPM7 may contribute directly to transepithelial Ca2+ transport in amelogenesis.


Asunto(s)
Ameloblastos/metabolismo , Calcio/metabolismo , Canales Catiónicos TRPM/metabolismo , Ameloblastos/citología , Ameloblastos/efectos de los fármacos , Anilidas/farmacología , Animales , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Incisivo/citología , Activación del Canal Iónico/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Mibefradil/farmacología , Ratones , Modelos Biológicos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Ratas , Tiadiazoles/farmacología
8.
Biochem Biophys Res Commun ; 525(4): 1011-1017, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32178872

RESUMEN

In seminiferous epithelium, tight junctions (TJs) between adjacent Sertoli cells constitute the blood-testis barrier and must change synchronically for germ cells to translocate from the basal to the adluminal compartment during the spermatogenic cycle. Rho GTPase activation through stimulation with specific L-selectin ligands has been proposed to modulate tight junctional dynamics. However, little is known regarding the role of Ca+2 dynamics in Sertoli cell and how Ca+2 relays L-selectin signals to modulate Rho GTPase activity in Sertoli cells, thus prompting us to investigate the Ca+2 flux induced by L-selectin ligand in ASC-17D cells. Using fluorescent real-time image, we first demonstrated the increase of intracellular Ca+2 level following L-selectin ligand stimulation. This Ca+2 increase was inhibited in ASC-17D cells pretreated with nifedipine, the L-type voltage-operated Ca+2 channel (VOCC) blocker, but not mibefradil, the T-type VOCC blocker. We then demonstrated the up-regulation of Rho and Rac1 in ASC-17D cells following the administration of L-selectin ligand, and the pre-treatment with nifedipine, but not mibefradil, prior to L-selectin ligand-binding abolished the activation of both Rho and Rac1. Together, we conclude that the activation of L-selectin induces Ca+2 influx through the L-type VOCC, which up-regulates Rho and Rac1 proteins, in ASC-17D cells.


Asunto(s)
Calcio/metabolismo , Selectina L/metabolismo , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio , Línea Celular , Ligandos , Masculino , Mibefradil/farmacología , Nifedipino/farmacología , Imagen Óptica , Ratas , Células de Sertoli/efectos de los fármacos , Células de Sertoli/enzimología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/genética
9.
Synapse ; 74(9): e22155, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32215948

RESUMEN

Epileptogenesis is a dynamical process that involves synaptic plasticity changes such as synaptic reorganization of excitatory and inhibitory systems and axonal sprouting in the hippocampus, which is one of the most studied epileptogenic regions in the brain. However, the early events that trigger these changes are not understood well. We investigated short-term and long-term synaptic plasticity parameters and T-type Ca2+ channel activity changes in the early phase of a rat kindling model. Chronic pentylenetetrazole (PTZ) application was used in order to induce the kindling process in rats. The recordings were obtained from hippocampal slices in the CA1 region at 25th day of PTZ application. Tetraethylammonium was used in order to induce long-term potentiation and T-type Ca2+ channel activity was assessed in the presence of mibefradil. We found that tetraethylammonium-induced long-term potentiation was not prevented by mibefradil in the kindling group in contrast to control group. We also found an increase in paired-pulse ratios in the PTZ-applied group. Our findings indicate an increase in the "T-type Ca2+ channel component of LTP" in the kindling group, which may be an early mechanism in epileptogenesis.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Canales de Calcio Tipo T/metabolismo , Epilepsia/metabolismo , Potenciación a Largo Plazo , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiopatología , Bloqueadores de los Canales de Calcio/farmacología , Epilepsia/etiología , Epilepsia/fisiopatología , Masculino , Mibefradil/farmacología , Pentilenotetrazol/toxicidad , Ratas , Ratas Wistar , Tetraetilamonio/farmacología
10.
J Cardiovasc Pharmacol ; 76(2): 246-254, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32433360

RESUMEN

Cardiac hypertrophy causes heart failure and is associated with hyperglycemia in patients with diabetes mellitus. Mibefradil, which acts as a T-type calcium channel blocker, exerts beneficial effects in patients with heart failure. In this study, we explored the effects and mechanism of mibefradil on high-glucose-induced cardiac hypertrophy in H9c2 cells. H9c2 cells were incubated in a high-glucose medium and then treated with different concentrations of mibefradil in the presence or absence of the Akt inhibitor MK2206 or mTOR inhibitor rapamycin. Cell size was evaluated through immunofluorescence, and mRNA expression of cardiac hypertrophy markers (atrial natriuretic peptide, brain natriuretic peptide, and ß-myosin heavy chain) was assessed by using quantitative real-time polymerase chain reaction. Changes in the expression of p-PI3K, p-Akt, and p-mTOR were evaluated using Western blotting, and autophagosome formation was detected using transmission electron microscopy. Our results indicate that mibefradil reduced the size of H9c2 cells, decreased mRNA expression of atrial natriuretic peptide, brain natriuretic peptide, and ß-myosin heavy chain, and decreased the level of autophagic flux. However, MK2206 and rapamycin induced autophagy and reversed the effects of mibefradil on high-glucose-induced H9c2 cells. In conclusion, mibefradil ameliorated high-glucose-induced cardiac hypertrophy by activating the PI3K/Akt/mTOR pathway and inhibiting excessive autophagy. Our study shows that mibefradil can be used therapeutically to ameliorate cardiac hypertrophy in patients with diabetes mellitus.


Asunto(s)
Autofagia/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Cardiomegalia/prevención & control , Glucosa/toxicidad , Mibefradil/farmacología , Miocitos Cardíacos/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Cardiomegalia/inducido químicamente , Cardiomegalia/enzimología , Cardiomegalia/patología , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/ultraestructura , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , Fosforilación , Ratas , Transducción de Señal
11.
Mediators Inflamm ; 2020: 3691701, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33223955

RESUMEN

Recent studies have illuminated that blocking Ca2+ influx into effector cells is an attractive therapeutic strategy for lung injury. We hypothesize that T-type calcium channel may be a potential therapeutic target for acute lung injury (ALI). In this study, the pharmacological activity of mibefradil (a classical T-type calcium channel inhibitor) was assessed in a mouse model of lipopolysaccharide- (LPS-) induced ALI. In LPS challenged mice, mibefradil (20 and 40 mg/kg) dramatically decreased the total cell number, as well as the productions of TNF-α and IL-6 in bronchoalveolar lavage fluid (BALF). Mibefradil also suppressed total protein concentration in BALF, attenuated Evans blue extravasation, MPO activity, and NF-κB activation in lung tissue. Furthermore, flunarizine, a widely prescripted antimigraine agent with potent inhibition on T-type channel, was also found to protect mice against lung injury. These data demonstrated that T-type calcium channel inhibitors may be beneficial for treating acute lung injury. The important role of T-type calcium channel in the acute lung injury is encouraged to be further investigated.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Flunarizina/farmacología , Lipopolisacáridos/metabolismo , Mibefradil/farmacología , Lesión Pulmonar Aguda/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/metabolismo , Citocinas/metabolismo , Pulmón/metabolismo , Lesión Pulmonar/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Prostate ; 79(13): 1580-1586, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31334879

RESUMEN

BACKGROUND: Androgen deprivation therapy (ADT) is the treatment of choice for metastatic prostate cancer (PCa). After an initial response to ADT, PCa cells can generate castration resistant (CRPC) or neuroendocrine (NEPC) malignancies, which are incurable. T-type calcium channels (TTCCs) are emerging as promising therapeutic targets for several cancers, but their role in PCa progression has never been investigated. METHODS: To examine the role of TTCCs in PCa, we analyzed their expression level, copy number variants (CNV) and prognostic significance using clinical datasets (Oncomine and cBioPortal). We then evaluated TTCC expression in a panel of PCa cell lines and measured the effect of their inhibition on cell proliferation and survival using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and caspase assays. RESULTS: TTCCs were upregulated in PCas harboring androgen receptor (AR) mutations; CNV rate was positively associated with PCa progression. Higher expression of one TTCC isoform (CACNA1G) predicted poorer postoperative prognosis in early stage PCa samples. Pharmacological or small interfering RNA (siRNA)-based inhibition of TTCCs caused a decrease in PC-3 cell survival and proliferation. CONCLUSIONS: Our results show that TTCCs are overexpressed in advanced forms of PCa and correlate with a poorer prognosis. TTCC inhibition reduces cell proliferation and survival, suggesting that there may be possible value in the therapeutic targeting of TTCCs in advanced PCa.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/deficiencia , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/biosíntesis , Línea Celular Tumoral , Proliferación Celular/fisiología , Etosuximida/farmacología , Humanos , Masculino , Mibefradil/farmacología , Terapia Molecular Dirigida , Células PC-3 , Pronóstico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Regulación hacia Arriba
13.
Reprod Fertil Dev ; 31(9): 1463-1472, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31030724

RESUMEN

The mechanism that causes the detachment of spermatozoa from the oviductal reservoir around the time of ovulation remains to be elucidated. Because the cumulus cells of the bovine oocyte are known to secrete progesterone (P4), and P4 has been shown to act upon cation channels of spermatozoa (CatSper) in human spermatozoa, it was hypothesised that P4 could induce hyperactivation due to an influx of extracellular calcium, and this would facilitate detachment of spermatozoa from oviductal epithelial cells. Therefore, this study aimed to investigate the role and mechanism of action of P4 in the release of spermatozoa from bovine oviduct epithelial cells (BOEC). Initial dose-response assessments on sperm hyperactivation determined the optimum concentration of P4 (10 nM), mibefradil (a non-specific Ca2+ channel antagonist; 5µM), NNC 55-0396 dihydrochloride (NNC; a CatSper antagonist; 2µM), mifepristone (a classical and membrane P4 receptor antagonist; 400nM) and AG205 (a membrane P4 receptor antagonist; 10µM). BOEC explants were incubated with frozen-thawed bovine spermatozoa for 30min, following which loosely bound spermatozoa were removed. Two experiments were completed. In Experiment 1, BOECs were treated for 30min with either no treatment, P4, NNC, mibefradil, P4+mibefradil, P4+NNC, P4+mibefradil+NNC or P4+EGTA. In Experiment 2, BOECs were treated for 30min with either no treatment, P4, mifepristone, AG205, mifepristone+AG205, P4+mifepristone, P4+AG205 or P4+mifepristone+AG205. The number of spermatozoa remaining bound per millimetre squared of BOEC explant was determined. Progesterone stimulated the release of bound spermatozoa from BOEC explants, whereas NNC, mibefradil and EGTA inhibited this release. The release of spermatozoa by P4 was inhibited in the presence of both mifepristone and AG205, whereas the combination of both had the greatest inhibitory action on P4 release of spermatozoa. These findings suggest the presence of a P4 membrane receptor on bovine spermatozoa and that P4-induced release of spermatozoa from BOECs is likely mediated by extracellular Ca2+.


Asunto(s)
Calcio/metabolismo , Células Epiteliales/efectos de los fármacos , Oviductos/efectos de los fármacos , Progesterona/farmacología , Receptores de Progesterona/metabolismo , Espermatozoides/efectos de los fármacos , Animales , Bencimidazoles/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Bovinos , Ciclopropanos/farmacología , Células Epiteliales/citología , Femenino , Antagonistas de Hormonas/farmacología , Masculino , Mibefradil/farmacología , Mifepristona/farmacología , Naftalenos/farmacología , Oviductos/citología , Receptores de Progesterona/antagonistas & inhibidores , Capacitación Espermática/efectos de los fármacos , Motilidad Espermática/efectos de los fármacos , Espermatozoides/metabolismo
14.
Int J Mol Sci ; 20(17)2019 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-31480231

RESUMEN

Human cytochrome P450 3A4 (CYP3A4) is the most important drug-metabolizing enzyme. Some drugs and natural compounds can act as suicide (mechanism-based) inactivators of CYP3A4, leading to unanticipated drug-drug interactions, toxicity and therapeutic failures. Despite significant clinical and toxicological implications, the mechanism-based inactivation remains incompletely understood. This study provides the first direct insights into the interaction of CYP3A4 with three suicide substrates: mibefradil, an antihypertensive drug quickly withdrawn from the market; a semi-synthetic antibiotic azamulin; and a natural furanocoumarin, 6',7'-dihydroxybergamottin. Novel structural findings help better understand the suicide substrate binding and inhibitory mechanism, and can be used to improve the predictability of the binding ability, metabolic sites and inhibitory/inactivation potential of newly developed drugs and other chemicals relevant to public health.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/metabolismo , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/metabolismo , Furocumarinas/química , Furocumarinas/metabolismo , Mibefradil/química , Mibefradil/metabolismo , Triazoles/química , Triazoles/metabolismo , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Especificidad por Sustrato
15.
J Cell Physiol ; 233(6): 4735-4747, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29135027

RESUMEN

The sperm acrosome reaction (AR), an essential event for mammalian fertilization, involves Ca2+ permeability changes leading to exocytosis of the acrosomal vesicle. The acrosome, an intracellular Ca2+ store whose luminal pH is acidic, contains hydrolytic enzymes. It is known that acrosomal pH (pHacr ) increases during capacitation and this correlates with spontaneous AR. Some AR inducers increase intracellular Ca2+ concentration ([Ca2+ ]i ) through Ca2+ release from internal stores, mainly the acrosome. Catsper, a sperm specific Ca2+ channel, has been suggested to participate in the AR. Curiously, Mibefradil and NNC55-0396, two CatSper blockers, themselves elevate [Ca2+ ]i by unknown mechanisms. Here we show that these compounds, as other weak bases, can elevate pHacr , trigger Ca2+ release from the acrosome, and induce the AR in both mouse and human sperm. To our surprise, µM concentrations of NNC55-0396 induced AR even in nominally Ca2+ free media. Our findings suggest that alkalization of the acrosome is critical step for Ca2+ release from the acrosome that leads to the acrosome reaction.


Asunto(s)
Reacción Acrosómica/efectos de los fármacos , Acrosoma/efectos de los fármacos , Bencimidazoles/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Ciclopropanos/farmacología , Mibefradil/farmacología , Naftalenos/farmacología , Acrosoma/metabolismo , Animales , Canales de Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones
16.
Artículo en Inglés | MEDLINE | ID: mdl-29684576

RESUMEN

Daphnia magna heartbeat is myogenic-originating within the animal's heart. However, the mechanism for this myogenic automaticity is unknown. The mechanism underlying the automaticity of vertebrate myogenic hearts involves cells (pacemaker cells), which have a distinct set of ion channels that include hyperpolarization activated cyclic nucleotide-gated (HCN) and T-type calcium ion channels. We hypothesized that these ion channels also underlie the automatic myogenic heartbeat of Daphnia magna. The drugs, ZD7288 and mibefradil dihydrochloride, block HCN and T-type calcium ion channels respectively. Application of these drugs, in separate experiments, show that they inhibit the heartbeat of Daphnia magna in a dose-dependent manner. Calculation of the percent difference between the heart rate of pretreatment (before drug application) and heart rate following drug application (post-treatment) allowed us to graph a dose-response curve for both ZD7288 and mibefradil, revealing that ZD7288 produces a greater effect on decreasing heart rate. This indicates the HCN ion channels play a foremost role in generating Daphnia magna heartbeat. Our results show conclusively that HCN and T-type calcium ion channels underlie the automatic myogenic heartbeat in Daphnia magna-and suggest a conserved mechanism for generating myogenic heartbeat within the animal kingdom. Thus, Daphnia magna represents a credible model system for further exploration of cardiac physiology.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/efectos de los fármacos , Cardiotónicos/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/efectos de los fármacos , Daphnia/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Mibefradil/farmacología , Pirimidinas/farmacología , Animales , Bloqueadores de los Canales de Calcio/administración & dosificación , Cardiotónicos/administración & dosificación , Relación Dosis-Respuesta a Droga , Mibefradil/administración & dosificación , Pirimidinas/administración & dosificación
17.
J Cell Physiol ; 232(8): 2019-2032, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27255432

RESUMEN

Several reports credit mibefradil with tumor suppressing properties arising from its known inhibition of Ca2+ currents. Given that mibefradil (Mb) is also known to inhibit K+ channels, we decided to study the interaction between this organic compound and the tumor-related Kv10.1 channel. Here we report that Mb modulates the gating of Kv10.1. Mb induces an apparent inactivation from both open and early closed states where the channels dwell at hyperpolarized potentials. Additionally, Mb accelerates the kinetics of current activation, in a manner that depends on initial conditions. Our observations suggest that Mb binds to the voltage sensor domain of Kv10.1 channels, thereby modifying the gating of the channels in a way that in some, but not all, aspects opposes to the gating effects exerted by divalent cations. J. Cell. Physiol. 232: 2019-2032, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Activación del Canal Iónico/efectos de los fármacos , Mibefradil/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Sitios de Unión , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Cinética , Potenciales de la Membrana , Mibefradil/metabolismo , Modelos Biológicos , Bloqueadores de los Canales de Potasio/metabolismo , Unión Proteica , Transfección
18.
Pflugers Arch ; 469(9): 1189-1202, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28429070

RESUMEN

Synchrony of spontaneous Ca2+ transients among venular mural cells (smooth muscle cells and pericytes) in visceral organs relies on the intercellular spread of L-type voltage-dependent Ca2+ channel (LVDCC)-dependent depolarisations. However, the mechanisms underlying the synchrony of spontaneous Ca2+ transients between arteriolar mural cells are less understood. The spontaneous intracellular Ca2+ dynamics of arteriolar mural cells in the rat rectal submucosa were visualised by Cal-520 Ca2+ imaging to analyse their synchrony. The mural cells in fine arterioles that had a rounded cell body with several extended processes developed spontaneous 'synchronous' Ca2+ transients arising from Ca2+ released from sarcoendoplasmic reticulum Ca2+ stores. Gap junction blockers (3 µM carbenoxolone, 10 µM 18ß-glycyrrhetinic acid), a Ca2+-activated Cl- channel (CaCC) blocker (100 µM 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid) or lowering extracellular Cl- concentration (from 134.4 to 12.4 mM) disrupted the synchrony of Ca2+ transients between arteriolar mural cells. Blockers of T-type voltage-dependent Ca2+ channels (TVDCCs, 1 µM mibefradil or ML218) or LVDCCs (1 µM nifedipine) reduced the Ca2+ transient frequency or their area under curve (AUC), respectively. However, neither TVDCC nor LVDCC blockers disrupted the synchrony of Ca2+ transients among arteriolar mural cells. This is in contrast with rectal venules in which nifedipine disrupted the synchrony of spontaneous Ca2+ transients. Thus, spontaneous transient depolarisations arising from the opening of CaCCs may effectively spread to neighbouring arteriolar mural cells via gap junctions to maintain the Ca2+ transient synchrony. Activation of TVDCCs appears to accelerate spontaneous Ca2+ transients, while LVDCCs predominantly contribute to the duration of Ca2+ transients.


Asunto(s)
Arteriolas/metabolismo , Calcio/metabolismo , Miocitos del Músculo Liso/metabolismo , Pericitos/metabolismo , Recto/metabolismo , Animales , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Canales de Cloruro/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Mibefradil/farmacología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Nifedipino/farmacología , Pericitos/efectos de los fármacos , Ratas , Ratas Wistar , Vénulas/metabolismo
19.
Biochem Biophys Res Commun ; 487(4): 801-806, 2017 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-28450109

RESUMEN

T-type calcium channels are prominently expressed in primary nociceptive fibers and well characterized in pain processes. Although itch and pain share many similarities including primary sensory fibers, the function of T-type calcium channels on acute itch has not been explored. We investigated whether T-type calcium channels expressed within primary sensory fibers of mouse skin, especially Cav3.2 subtype, involve in chloroquine-, endothelin-1- and histamine-evoked acute itch using pharmacological, neuronal imaging and behavioral analyses. We found that pre-locally blocking three subtypes of T-type calcium channels in the peripheral afferents of skins, yielded an inhibition in acute itch or pain behaviors, while selectively blocking the Cav3.2 channel in the skin peripheral afferents only inhibited acute pain but not acute itch. These results suggest that T-type Cav3.1 or Cav3.3, but not Cav3.2 channel, have an important role in acute itch processing, and their distinctive roles in modulating acute itch are worthy of further investigation.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Neuronas Aferentes/metabolismo , Prurito/metabolismo , Piel/metabolismo , Animales , Masculino , Mibefradil/farmacología , Ratones , Ratones Endogámicos C57BL
20.
Nature ; 471(7338): 382-6, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21412338

RESUMEN

In the oviduct, cumulus cells that surround the oocyte release progesterone. In human sperm, progesterone stimulates a Ca(2+) increase by a non-genomic mechanism. The Ca(2+) signal has been proposed to control chemotaxis, hyperactivation and acrosomal exocytosis of sperm. However, the underlying signalling mechanism has remained mysterious. Here we show that progesterone activates the sperm-specific, pH-sensitive CatSper Ca(2+) channel. We found that both progesterone and alkaline pH stimulate a rapid Ca(2+) influx with almost no latency, incompatible with a signalling pathway involving metabotropic receptors and second messengers. The Ca(2+) signals evoked by alkaline pH and progesterone are inhibited by the Ca(v) channel blockers NNC 55-0396 and mibefradil. Patch-clamp recordings from sperm reveal an alkaline-activated current carried by mono- and divalent ions that exhibits all the hallmarks of sperm-specific CatSper Ca(2+) channels. Progesterone substantially enhances the CatSper current. The alkaline- and progesterone-activated CatSper current is inhibited by both drugs. Our results resolve a long-standing controversy over the non-genomic progesterone signalling. In human sperm, either the CatSper channel itself or an associated protein serves as the non-genomic progesterone receptor. The identification of CatSper channel blockers will greatly facilitate the study of Ca(2+) signalling in sperm and help to define further the physiological role of progesterone and CatSper.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Progesterona/farmacología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Alprostadil/farmacología , Bencimidazoles/farmacología , Bloqueadores de los Canales de Calcio/farmacología , AMP Cíclico , Ciclopropanos/farmacología , Conductividad Eléctrica , Humanos , Concentración de Iones de Hidrógeno , Masculino , Mibefradil/farmacología , Naftalenos/farmacología , Técnicas de Placa-Clamp , Progesterona/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda