Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Physiol ; 601(14): 2935-2958, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37278367

ABSTRACT

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.


Subject(s)
Calcium , Semen , Male , Animals , Mice , Calcium/metabolism , Semen/metabolism , Spermatozoa/metabolism , Acrosome/metabolism , Mibefradil/metabolism , Mibefradil/pharmacology , Hydrogen-Ion Concentration , Mammals/metabolism
2.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555708

ABSTRACT

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.


Subject(s)
Kidney , Nifedipine , Receptor, Notch3 , Animals , Mice , Angiotensin II/pharmacology , Arterioles/metabolism , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Kidney/metabolism , Mibefradil/metabolism , Nifedipine/pharmacology , Vascular Resistance , Receptor, Notch3/genetics , Gene Deletion , Mice, Knockout
3.
Nan Fang Yi Ke Da Xue Xue Bao ; 42(7): 1032-1037, 2022 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-35869766

ABSTRACT

OBJECTIVE: To observe the effect of mibefradil on skeletal muscle mass, function and structure in obese mice. METHODS: Fifteen 6-week-old C57BL/6 mice were randomized equally into normal diet group (control group), high-fat diet (HFD) group and high-fat diet +mibefradil intervention group (HFD +Mibe group). The grip strength of the mice was measured using an electronic grip strength meter, and the muscle content of the hindlimb was analyzed by X-ray absorptiometry (DXA). Triglyceride (TG) and total cholesterol (TC) levels of the mice were measured with GPO-PAP method. The cross-sectional area of the muscle fibers was observed with HE staining. The changes in the level of autophagy in the muscles were detected by Western blotting and immunofluorescence assay, and the activation of the Akt/mTOR signaling pathway was detected with Western blotting. RESULTS: Compared with those in the control group, the mice in HFD group had a significantly greater body weight, lower relative grip strength, smaller average cross sectional area of the muscle fibers, and a lower hindlimb muscle ratio (P < 0.05). Immunofluorescence assay revealed a homogenous distribution of LC3 emitting light red fluorescence in the cytoplasm in the muscle cells in HFD group and HFD+Mibe group, while bright spots of red fluorescence were detected in HFD group. In HFD group, the muscular tissues of the mice showed an increased expression level of LC3 II protein with lowered expressions of p62 protein and phosphorylated AKT and mTOR (P < 0.05). Mibefradil treatment significantly reduced body weight of the mice, lowered the expression level of p62 protein, and increased forelimb grip strength, hindlimb muscle ratio, cross-sectional area of the muscle fibers, and the expression levels of LC3 II protein and phosphorylated AKT and mTOR (P < 0.05). CONCLUSION: Mibefradil treatment can moderate high-fat diet-induced weight gain and improve muscle mass and function in obese mice possibly by activating AKT/mTOR signal pathway to improve lipid metabolism and inhibit obesityinduced autophagy.


Subject(s)
Diet, High-Fat , Proto-Oncogene Proteins c-akt , Animals , Body Weight , Mibefradil/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
4.
BMC Nephrol ; 23(1): 211, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35710406

ABSTRACT

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.


Subject(s)
Calcium Channels, T-Type , Mesangial Cells , Animals , Humans , Mesangial Cells/metabolism , Mibefradil/metabolism , Mibefradil/pharmacology , Mice , Phosphorylation , Rats , Transforming Growth Factor beta1/metabolism
5.
Int Arch Allergy Immunol ; 183(6): 579-590, 2022.
Article in English | MEDLINE | ID: mdl-35100604

ABSTRACT

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.


Subject(s)
Calcium Channels, T-Type , Cilia , Adenosine Triphosphate/metabolism , Calcium/metabolism , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cilia/physiology , Cyclic GMP , Epithelial Cells/metabolism , Humans , Mibefradil/metabolism , Mibefradil/pharmacology , Nasal Mucosa/metabolism
6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-941037

ABSTRACT

OBJECTIVE@#To observe the effect of mibefradil on skeletal muscle mass, function and structure in obese mice.@*METHODS@#Fifteen 6-week-old C57BL/6 mice were randomized equally into normal diet group (control group), high-fat diet (HFD) group and high-fat diet +mibefradil intervention group (HFD +Mibe group). The grip strength of the mice was measured using an electronic grip strength meter, and the muscle content of the hindlimb was analyzed by X-ray absorptiometry (DXA). Triglyceride (TG) and total cholesterol (TC) levels of the mice were measured with GPO-PAP method. The cross-sectional area of the muscle fibers was observed with HE staining. The changes in the level of autophagy in the muscles were detected by Western blotting and immunofluorescence assay, and the activation of the Akt/mTOR signaling pathway was detected with Western blotting.@*RESULTS@#Compared with those in the control group, the mice in HFD group had a significantly greater body weight, lower relative grip strength, smaller average cross sectional area of the muscle fibers, and a lower hindlimb muscle ratio (P < 0.05). Immunofluorescence assay revealed a homogenous distribution of LC3 emitting light red fluorescence in the cytoplasm in the muscle cells in HFD group and HFD+Mibe group, while bright spots of red fluorescence were detected in HFD group. In HFD group, the muscular tissues of the mice showed an increased expression level of LC3 II protein with lowered expressions of p62 protein and phosphorylated AKT and mTOR (P < 0.05). Mibefradil treatment significantly reduced body weight of the mice, lowered the expression level of p62 protein, and increased forelimb grip strength, hindlimb muscle ratio, cross-sectional area of the muscle fibers, and the expression levels of LC3 II protein and phosphorylated AKT and mTOR (P < 0.05).@*CONCLUSION@#Mibefradil treatment can moderate high-fat diet-induced weight gain and improve muscle mass and function in obese mice possibly by activating AKT/mTOR signal pathway to improve lipid metabolism and inhibit obesityinduced autophagy.


Subject(s)
Animals , Mice , Body Weight , Diet, High-Fat , Mibefradil/metabolism , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
7.
Int J Mol Sci ; 20(17)2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31480231

ABSTRACT

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.


Subject(s)
Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/metabolism , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Furocoumarins/chemistry , Furocoumarins/metabolism , Mibefradil/chemistry , Mibefradil/metabolism , Triazoles/chemistry , Triazoles/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Substrate Specificity
8.
J Cell Physiol ; 232(8): 2019-2032, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27255432

ABSTRACT

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.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ion Channel Gating/drug effects , Mibefradil/pharmacology , Potassium Channel Blockers/pharmacology , Binding Sites , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Kinetics , Membrane Potentials , Mibefradil/metabolism , Models, Biological , Potassium Channel Blockers/metabolism , Protein Binding , Transfection
9.
Bioorg Med Chem Lett ; 21(19): 5910-5, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21843937

ABSTRACT

To obtain selective and potent inhibitor for T-type calcium channel by ligand based drug design, 4-piperidinecarboxylate and 4-piperidinecyanide derivatives were prepared and evaluated for in vitro and in vivo activity against α(1G) calcium channel. Among them, several compounds showed good T-type calcium channel inhibitory activity and minimal off-target activity over hERG channel (% inhibition at 10 µM=61.85-71.99, hERG channel IC(50)=1.57 ± 0.14-4.98 ± 0.36 µM). Selected compound 31a was evaluated on SNL model of neuropathic pain and showed inhibitory effect on mechanical allodynia.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Animals , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/metabolism , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Drug Stability , HEK293 Cells , Humans , Hyperalgesia/physiopathology , Inhibitory Concentration 50 , Ligands , Mibefradil/metabolism , Molecular Structure , Molecular Targeted Therapy , Neuralgia/physiopathology , Patch-Clamp Techniques , Piperidines/chemistry , Piperidines/metabolism , Quantitative Structure-Activity Relationship , Rats , Spinal Nerves/surgery , Structure-Activity Relationship
10.
Fundam Clin Pharmacol ; 25(4): 469-78, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21039820

ABSTRACT

Recent in vitro evidence suggests that T-type Ca(2+) channels are implicated in the mechanisms of ischemia-induced delayed neuronal cell death. The aim of this work was to study the neuroprotective potential of mibefradil and pimozide, both T-type Ca(2+) channel inhibitors, in an in vivo rat model of global ischemia. We performed blinded and randomized placebo vs. treatment experiments using 57 animals to test mibefradil and fourteen animals to test pimozide. Each treated animal received a single stereotactic intraventricular injection of mibefradil or intraperitoneal injection of pimozide prior to transient global cerebral ischemia. The primary endpoint was the number of neurons surviving in the CA1 region 72 h after insult as evaluated by NeuN-labeled cell counts. All physiological variables monitored immediately before and after ischemic insult were equivalent between all groups. Surviving neurons in the CA1 region were significantly more frequent in the treated groups compared to the placebo group (mibefradil: 36.8 ± 2.8 cells in a 200 × 100 µm counting area vs. placebo: 25.2 ± 3.2 [P < 0.01]; pimozide: 39.4 ± 1.12 vs. placebo: 27.8 ± 0.7 [P < 0.0001]). Thus, administration of mibefradil or pimozide effectively prevents neuronal death after ischemia in a rat model of global ischemia. This study provides further support for a neuroprotective effect of T-type Ca(2+) current inhibition during ischemia.


Subject(s)
CA1 Region, Hippocampal/drug effects , Calcium Channel Blockers/therapeutic use , Ischemic Attack, Transient/drug therapy , Mibefradil/therapeutic use , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Pimozide/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , CA1 Region, Hippocampal/pathology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Glucose/deficiency , HEK293 Cells , Humans , Hypoxia , Ischemic Attack, Transient/blood , Ischemic Attack, Transient/mortality , Ischemic Attack, Transient/pathology , Lactic Acid/blood , Male , Membrane Potentials/physiology , Mibefradil/metabolism , Mibefradil/pharmacokinetics , Mibefradil/pharmacology , Neurons/pathology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Pimozide/pharmacology , Rats , Rats, Sprague-Dawley , Survival Analysis , Tissue Culture Techniques , Transfection
11.
Am J Physiol Lung Cell Mol Physiol ; 299(1): L86-97, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20435690

ABSTRACT

Regulated P-selectin surface expression provides a rapid measure for endothelial transition to a proinflammatory phenotype. In general, P-selectin surface expression results from Weibel-Palade body (WPb) exocytosis. Yet, it is unclear whether pulmonary capillary endothelium possesses WPbs or regulated P-selectin surface expression and, if so, how inflammatory stimuli initiate exocytosis. We used immunohistochemistry, immunofluorescence labeling, ultrastructural assessment, and an isolated perfused lung model to demonstrate that capillary endothelium lacks WPbs but possesses P-selectin. Thrombin stimulated P-selectin surface expression in both extra-alveolar vessel and alveolar capillary endothelium. Only in capillaries was the thrombin-stimulated P-selectin surface expression considerably mitigated by pharmacologic blockade of the T-type channel or genetic knockout of the T-type channel alpha(1G)-subunit. Depolarization of endothelial plasma membrane via high K(+) perfusion capable of eliciting cytosolic Ca(2+) transients also provoked P-selectin surface expression in alveolar capillaries that was abolished by T-type channel blockade or alpha(1G) knockout. Our findings reveal an intracellular WPb-independent P-selectin pool in pulmonary capillary endothelium, where the regulated P-selectin surface expression is triggered by Ca(2+) transients evoked through activation of the alpha(1G) T-type channel.


Subject(s)
Calcium Channels, T-Type/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Lung/blood supply , P-Selectin/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, T-Type/genetics , Calcium Signaling/physiology , Endothelial Cells/ultrastructure , Endothelium, Vascular/ultrastructure , Exocytosis/physiology , Humans , Lung/ultrastructure , Male , Mibefradil/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Weibel-Palade Bodies/metabolism , Weibel-Palade Bodies/ultrastructure , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
12.
J Biol Chem ; 285(10): 7447-58, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20056611

ABSTRACT

Here we describe features of the first non-mammalian T-type calcium channel (LCa(v)3) expressed in vitro. This molluscan channel possesses combined biophysical properties that are reminiscent of all mammalian T-type channels. It exhibits T-type features such as "transient" kinetics, but the "tiny" label, usually associated with Ba(2+) conductance, is hard to reconcile with the "bigness" of this channel in many respects. LCa(v)3 is 25% larger than any voltage-gated ion channel expressed to date. It codes for a massive, 322-kDa protein that conducts large macroscopic currents in vitro. LCa(v)3 is also the most abundant Ca(2+) channel transcript in the snail nervous system. A window current at typical resting potentials appears to be at least as large as that reported for mammalian channels. This distant gene provides a unique perspective to analyze the structural, functional, drug binding, and evolutionary aspects of T-type channels.


Subject(s)
Calcium Channels, T-Type/metabolism , Central Nervous System/metabolism , Ion Channel Gating/physiology , Lymnaea , Amino Acid Sequence , Animals , Base Sequence , Calcium Channel Blockers/metabolism , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/classification , Calcium Channels, T-Type/genetics , Cell Line , Humans , Lymnaea/anatomy & histology , Lymnaea/metabolism , Mibefradil/metabolism , Molecular Sequence Data , Nickel/metabolism , Patch-Clamp Techniques , Phylogeny , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid
13.
Bioorg Med Chem ; 14(10): 3502-11, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16434203

ABSTRACT

For the novel, potent, and selective T-type Ca2+ channel blockers, a series of sulfonamido-containing 3,4-dihydroquinazoline derivatives were prepared and evaluated for their blocking actions on T- and N-type Ca2+ channels. Among them, 9c (KYS05064, IC50 = 0.96 +/- 0.22 microM) was found to be as potent as Mibefradil and also showed the highest selectivity for T-type Ca2+ channel with no effect on N-type Ca2+ channel.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Calcium Channels, T-Type/drug effects , Quantitative Structure-Activity Relationship , Calcium Channel Blockers/chemical synthesis , Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/metabolism , Cell Line , Cells, Cultured , Electrophysiology , Humans , Lethal Dose 50 , Mibefradil/chemistry , Mibefradil/metabolism , Mibefradil/pharmacology , Molecular Structure , Quinazolines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacology , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology
14.
Toxicol Pathol ; 33(3): 356-64, 2005.
Article in English | MEDLINE | ID: mdl-15805073

ABSTRACT

An unexpected dose related increase in oral squamous cell carcinomas was observed in a standard 2-year carcinogenicity study with a novel calcium channel blocker, in which Wistar rats received daily doses of 0, 1.5, 7, 20, or 40 mg/kg of the compound mixed with a standard diet containing fibers from barley. This finding was associated with an increased incidence of severe (destructive) periodontitis and the formation of oro-nasal fistulae at the 2 highest doses. Five assays of the compound for genotoxicity were negative indicating that a genotoxic effect was highly improbable. To investigate the underlying pathogenic mechanisms a second 2-year study in the same strain of rats was initiated and the influence of the diet and/or a possible local irritancy by the drug was assessed. In this second study the compound was administered by oral gavage at daily doses of 0, 7, or 40 mg/kg (later reduced to 20 mg/kg due to systemic intolerance) to rats maintained either on the standard diet or on a low fiber diet assumed to be less aggressive in terms of inducing periodontal lesions. Dose dependent gingival overgrowth (a class-related effect) was observed in the incisor and molar teeth area of all treated groups but was independent of the diet used. No oral tumors were found in the standard diet or low fiber diet controls and all treatment groups fed the low fiber diet, whereas in the high-dose group fed the standard diet a total of 8 oral squamous cell carcinomas were detected in association with an increased incidence of severe periodontitis. These results indicate that the increased incidence of squamous cell carcinomas observed upon chronic administration of the compound is not due to a direct tumorigenic effect of the drug. Tumor formation is attributable to severe periodontal disease favored by the diet and class related gingival overgrowth.


Subject(s)
Calcium Channel Blockers/toxicity , Calcium Channels/drug effects , Carcinoma, Squamous Cell/metabolism , Diet , Mouth Neoplasms/metabolism , Administration, Oral , Animals , Calcium Channel Blockers/metabolism , Calcium Channels/metabolism , Carcinoma, Squamous Cell/pathology , Dose-Response Relationship, Drug , Female , Male , Mibefradil/chemistry , Mibefradil/metabolism , Mibefradil/toxicity , Molecular Structure , Mouth Neoplasms/pathology , Rats , Rats, Wistar , Toxicity Tests, Chronic
15.
Biochem Biophys Res Commun ; 324(1): 401-8, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15465033

ABSTRACT

In order to investigate the currently unknown cellular signaling pathways of T-type Ca(2+) channels, we decided to construct a new cell line which would stably express alpha(1G) and Kir2.1 subunits in HEK293 cells (HEK293/alpha(1G)/Kir2.1). Compared to cells which only expressed alpha(1G) (HEK293/alpha(1G)), HEK293/alpha(1G)/Kir2.1 cells produced an enormous inward rectifying current which was blocked by external Ba(2+) and Cs(+) in a concentration-dependent manner. The expression of Kir2.1 channels contributed significantly to the shift of membrane potential from -12.2+/-2.8 to -57.3+/-3.7mV. However, biophysical and pharmacological properties of alpha(1G)-mediated Ca(2+) channels remained unaffected by the expression of Kir2.1 subunits, except for the enlarging of the window current region. Biochemical activation of alpha(1G) channels using 150mM KCl brought about an increase in [Ca(2+)](i), which was blocked by mibefradil, the T-type Ca(2+) channel blocker. These data suggest that the HEK293/alpha(1G)/Kir2.1 cell line would have potential uses in the study of T-type Ca(2)(+) channel-mediated signaling pathways and possibly useful in the development of new therapeutic drugs associated with T-type Ca(2)(+) channels.


Subject(s)
Calcium Channels, T-Type/metabolism , Cell Line , Kidney , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/metabolism , Signal Transduction/physiology , Barium/metabolism , Calcium/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, T-Type/genetics , Cesium/metabolism , Humans , Kidney/cytology , Kidney/embryology , Membrane Potentials/physiology , Mibefradil/metabolism , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Protein Subunits/genetics , Transfection
16.
Membr Cell Biol ; 13(5): 645-55, 2000.
Article in English | MEDLINE | ID: mdl-10987388

ABSTRACT

Here we report that a Ca2+ antagonist mibefradil (Ro 40-5967) which has been shown to be a selective inhibitor of T-type calcium channels increases free calcium concentration ([Ca2+]i) in the cytoplasm of cultured smooth muscle cells isolated from porcine coronary artery. Smooth muscle cells were loaded with Fura 2 and a videoimage system was used to follow the [Ca2+]i responses. It was shown that at a concentration of 1 nM mibefradil induced a transient [Ca2+]i elevation in individual cells and at a concentration of 100 nM this compound stimulated almost all the cells in monolayer. The [Ca2+]i response did not change with the further increase of the mibefradil concentration up to 10 microM. The half-maximal effect was observed at 10 nM. The increase in [Ca2+]i strongly depended on the presence of Ca in the extracellular medium. Calcium antagonists belonging to three different classes--verapamil (phenylalkylamines), diltiazem (benzothiazepines) and amlodipin (dihydropyridines) neither suppressed the mibefradil effect nor mimicked it. These data indicate that mibefradil increased [Ca2+]i acting via a distinct receptor site. We suggest that these receptors are coupled to calcium channels of plasma membrane.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channels, T-Type/physiology , Mibefradil/metabolism , Amlodipine/metabolism , Amlodipine/pharmacology , Animals , Calcium/agonists , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Coronary Vessels/cytology , Diltiazem/metabolism , Diltiazem/pharmacology , Mibefradil/pharmacology , Muscle, Smooth, Vascular/cytology , Swine , Verapamil/metabolism , Verapamil/pharmacology
17.
Naunyn Schmiedebergs Arch Pharmacol ; 361(6): 578-83, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10882031

ABSTRACT

Briefly after withdrawal of the (T-type) calcium channel blocker mibefradil from the market, four cases of life-threatening interaction of mibefradil with dihydropyridines were reported. We investigated in vitro whether mibefradil interacts with a dihydropyridine, as described for other non-dihydropyridine compounds. Rat working hearts were used to examine functional interactions between amlodipine and mibefradil. Gallopamil and another T-type-channel blocker, ethosuximide, were included for comparison. Effects of mibefradil, (+)- and (-)-gallopamil on [3H](+)-isradipine binding were studied in membranes from tsA201-cells transfected with alpha(1c)-, alpha(2)delta-, and beta(1a)- or beta(2a)-calcium channel subunits. Mibefradil increased negative inotropic effect of amlodipine, but not of gallopamil. Gallopamil and ethosuximide showed no influence on contractile effects of amlodipine. Furthermore, mibefradil concentration-dependently caused bradycardic rhythm disturbance. The same type of arrhythmia was observed combining low concentrations of mibefradil with amlodipine, or with gallopamil, respectively. Amlodipine alone, or the combination of gallopamil or ethosuximide with amlodipine did not cause any arrhythmia. Binding studies showed a concentration-dependent positive allosteric interaction between [3H](+)-isradipine and mibefradil, but not with [3H](+)-isradipine and gallopamil enantiomers. Molecular and functional evidence points to an interaction between a dihydropyridine and mibefradil. Mibefradil caused rhythm disturbances and potentiation of negative inotropy when combined with amlodipine.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Dihydropyridines/pharmacology , Mibefradil/pharmacology , Myocardial Contraction/drug effects , Ventricular Pressure/drug effects , Amlodipine/pharmacology , Animals , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/genetics , Cell Line , Cell Membrane/metabolism , Dihydropyridines/metabolism , Drug Interactions , Ethosuximide/pharmacology , Female , Gallopamil/pharmacology , Humans , In Vitro Techniques , Isradipine/metabolism , Male , Mibefradil/metabolism , Perfusion , Radioligand Assay , Rats , Rats, Wistar
18.
Drug Metab Dispos ; 28(8): 895-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10901697

ABSTRACT

Mibefradil, a calcium T- and L-channel blocker developed for use in hypertension, was recently removed from the market after reports of severe drug-drug interactions. Mibefradil is known to inhibit various cytochrome P450 enzymes involved in drug metabolism, particularly CYP3A. However, the extent and the severity of the observed drug interactions in humans suggest that inhibition of additional systems important to drug disposition, such as the drug transporter P-glycoprotein (P-gp), may also have contributed to the severity of the mibefradil interactions. A polarized epithelial cell line, LLC-PK1, which does not express P-gp, and the derived L-MDR1 cell line, which overexpresses human P-gp, were used to study the effects of mibefradil on drug transport. A markedly greater basal-to-apical versus apical-to-basal transport of [H3]mibefradil was seen in the L-MDR1, but not in the LLC-PK1 cells, suggesting that the drug is a substrate of P-gp. Using a human intestinal cancer-derived cell line Caco-2, which constitutively expresses P-gp, mibefradil was shown to be a potent inhibitor of P-gp-mediated digoxin transport, with an IC50 of 1.6 microM. Additionally, the effect of mibefradil on CYP3A was assessed using human liver microsomes. Mibefradil inhibited CYP3A-mediated nifedipine oxidase activity with an IC50 of 0.8 microM, and a Ki of 0.6 microM. Thus, mibefradil is not only a P-gp substrate, but also a potent inhibitor of both P-gp and CYP3A. These data suggest that the severity of drug interactions seen with mibefradil use is due to the dual inhibition of both P-gp and CYP3A.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Mibefradil/pharmacology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Biological Transport/drug effects , Caco-2 Cells , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Cytochrome P-450 CYP3A , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Humans , Mibefradil/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism
19.
J Pharmacol Exp Ther ; 292(3): 939-43, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10688607

ABSTRACT

It has been shown that mibefradil (Ro 40-5967) exerts a selective inhibitory effect on T-type Ca(2+) currents, although at higher concentrations it can antagonize high voltage-activated Ca(2+) currents. The action of mibefradil on Ca(2+) channels is use- and steady-state-dependent and the binding site of mibefradil on L-type Ca(2+) channels is different from that of dihydropyridines. By using conventional whole-cell and perforated patch-clamp techniques, we showed that mibefradil has an inhibitory effect on both T- and L-type Ca(2+) currents in insulin-secreting cells. However, the effect on L-type Ca(2+) currents was time-dependent and poorly reversible in perforated patch-clamp experiments. By using mass spectrometry, we demonstrated that mibefradil accumulates inside cells, and furthermore, a metabolite of mibefradil was detected. Intracellular application of this metabolite selectively blocked the L-type Ca(2+) current, whereas mibefradil exerted no effect. This study demonstrates that mibefradil permeates into cells and is hydrolyzed to a metabolite that blocks L-type Ca(2+) channels specifically by acting at the inner side of the channel.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Islets of Langerhans/drug effects , Mibefradil/pharmacology , Calcium Channels, T-Type/drug effects , Cell Line , Mibefradil/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...