Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Mol Neurobiol ; 59(5): 2932-2945, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35243582

RESUMO

Medulloblastoma (MB) is the most common malignant paediatric brain tumour. In our previous studies, we developed a novel 3D assay for MB cells that was used to screen a panel of plasma membrane calcium channel modulators for their effect on the 3D growth of D341 MB cells. These studies identified T-type (CaV3) channel inhibitors, mibefradil and NNC-55-0396 (NNC) as selective inhibitors of MB cell growth. Mibefradil was originally approved for the treatment of hypertension and angina pectoris, and recently successfully completed a phase I trial for recurrent high-grade glioma. NNC is an analogue of mibefradil with multiple advantages compared to mibefradil that makes it attractive for potential future clinical trials. T-type channels have a unique low voltage-dependent activation/inactivation, and many studies suggest that they have a direct regulatory role in controlling Ca2+ signalling in non-excitable tissues, including cancers. In our previous study, we also identified overexpression of CaV3.2 gene in MB tissues compared to normal brain tissues. In this study, we aimed to characterise the effect of mibefradil and NNC on MB cells and elucidate their mechanism of action. This study demonstrates that the induction of toxicity in MB cells is selective to T-type but not to L-type Ca2+ channel inhibitors. Addition of CaV3 inhibitors to vincristine sensitised MB cells to this MB chemotherapeutic agent, suggesting an additive effect. Furthermore, CaV3 inhibitors induced cell death in MB cells via apoptosis. Supported by proteomics data and cellular assays, apoptotic cell death was associated with reduced mitochondrial membrane potential and reduced ATP levels, which suggests that both compounds alter the metabolism of MB cells. This study offers new insights into the action of mibefradil and NNC and will pave the way to test these molecules or their analogues in pre-clinical MB models alone and in combination with vincristine to assess their suitability as a potential MB therapy.


Assuntos
Canais de Cálcio Tipo T , Neoplasias Cerebelares , Meduloblastoma , Apoptose , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/metabolismo , Criança , Humanos , Meduloblastoma/tratamento farmacológico , Mibefradil/farmacologia , Mibefradil/uso terapêutico , Recidiva Local de Neoplasia , Vincristina/farmacologia
2.
Mol Med Rep ; 16(3): 3169-3178, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713955

RESUMO

Hippocampal neurogenesis in temporal lobe epilepsy (TLE) may result in alteration of the excitability of neurons, which contributes to spontaneous recurrent seizures. Axon initial segment (AIS) structural and functional plasticity is important in the control of neuronal excitability. It remains to be elucidated whether the plasticity of AIS occurs in hippocampal newly­generated neurons that are involved in recurrent seizures following pilocarpine­induced status epilepticus (SE). The present study first established a pilocarpine­induced TLE rat model to assess the features of newborn neurons and AIS plasticity alterations using double immunofluorescence staining of Ankyrin G and doublecortin (DCX). AIS plasticity alterations include length and distance from soma in the hippocampal newly­generated neurons post­SE. The results of the present study demonstrated that pilocarpine­induced epileptic rats exhibited aberrant hippocampal neurogenesis and longer DCX­labeled cell dendrites in the dentate gyrus. Pilocarpine­induced epileptic rats demonstrated shortened lengths of AIS and an increased distance from the soma in hippocampal newborn neurons. Mibefradil, a T/L­type calcium blocker, reversed the alterations in length and position of AIS in hippocampal newborn neurons post­SE, accompanied by decreased long­term seizure activity without increased aberrant neurogenesis. These findings indicate that the plasticity of AIS in hippocampal neurogenesis may have profound consequences in epilepsy, at least in animals.


Assuntos
Segmento Inicial do Axônio/patologia , Epilepsia do Lobo Temporal/patologia , Hipocampo/patologia , Neurogênese , Animais , Segmento Inicial do Axônio/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Dendritos/efeitos dos fármacos , Dendritos/patologia , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Proteína Duplacortina , Eletroencefalografia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Masculino , Mibefradil/farmacologia , Mibefradil/uso terapêutico , Neurogênese/efeitos dos fármacos , Pilocarpina , Ratos Sprague-Dawley , Recidiva
3.
Assay Drug Dev Technol ; 13(10): 650-3, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26690767

RESUMO

The expanding "valley of death" in drug development is leaving potentially life-saving new chemical entities and molecular targets fallow. This situation is forcing early-stage companies to think creatively about moving their technologies forward, especially as institutional investors show more interest in later stages of development. Drug repurposing, a strategy to examine existing drugs for therapeutic value against different diseases, is an emerging method to bring an off-market drug back onto the market. Tau Therapeutics LLC identified the role of T-type calcium channel blockers (Cav3) in cancer proliferation, but the company was unable to attract funding while having both a nonvalidated drug target and new chemical entities. To change the risk profile of the company, Tau set out to repurpose the known Cav3 drug mibefradil as a proof of concept for the treatment of cancer. Mibefradil was launched for hypertension in the 1990s but withdrawn because of drug-drug interactions. A new sequential combination treatment, termed Interlaced Therapy™, uses short-term administration of mibefradil to enhance the overall therapeutic potential of conventional anticancer agents. Mibefradil is currently in a phase Ib clinical trial with the National Cancer Institute (NCI) Adult Brain Tumor Consortium. Mibefradil has been repurposed from an abandoned antihypertensive to a targeted solid tumor treatment, and it has been rescued from drug-drug interactions by using short-term dose exposure. Tau is using the early success of mibefradil as a proof of concept to build a platform technology of Cav3 blockers for broad antitumor applications in combination with new targeted cancer therapies, well-established chemotherapies, and radiation.


Assuntos
Anti-Hipertensivos/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Reposicionamento de Medicamentos/métodos , Mibefradil/uso terapêutico , Neoplasias/tratamento farmacológico , Anti-Hipertensivos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/metabolismo , Ensaios Clínicos Fase I como Assunto , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Mibefradil/farmacologia
4.
PLoS One ; 10(5): e0127572, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25974104

RESUMO

The Cav3.2 isoform of the T-type calcium channel is expressed in primary sensory neurons of the dorsal root ganglion (DRG), and these channels contribute to nociceptive and neuropathic pain in rats. However, there are conflicting reports on the roles of these channels in pain processing in rats and mice. In addition, the function of T-type channels in persistent inflammatory hyperalgesia is poorly understood. We performed behavioral and comprehensive histochemical analyses to characterize Cav3.2-expressing DRG neurons and examined the regulation of T-type channels in DRGs from C57BL/6 mice with carrageenan-induced inflammatory hyperalgesia. We show that approximately 20% of mouse DRG neurons express Cav3.2 mRNA and protein. The size of the majority of Cav3.2-positive DRG neurons (69 ± 8%) ranged from 300 to 700 µm2 in cross-sectional area and 20 to 30 µm in estimated diameter. These channels co-localized with either neurofilament-H (NF-H) or peripherin. The peripherin-positive cells also overlapped with neurons that were positive for isolectin B4 (IB4) and calcitonin gene-related peptide (CGRP) but were distinct from transient receptor potential vanilloid 1 (TRPV1)-positive neurons during normal mouse states. In mice with carrageenan-induced inflammatory hyperalgesia, Cav3.2 channels, but not Cav3.1 or Cav3.3 channels, were upregulated in ipsilateral DRG neurons during the sub-acute phase. The increased Cav3.2 expression partially resulted from an increased number of Cav3.2-immunoreactive neurons; this increase in number was particularly significant for TRPV1-positive neurons. Finally, preceding and periodic intraplantar treatment with the T-type calcium channel blockers mibefradil and NNC 55-0396 markedly reduced and reversed mechanical hyperalgesia during the acute and sub-acute phases, respectively, in mice. These data suggest that Cav3.2 T-type channels participate in the development of inflammatory hyperalgesia, and this channel might play an even greater role in the sub-acute phase of inflammatory pain due to increased co-localization with TRPV1 receptors compared with that in the normal state.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Neurônios/metabolismo , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Canais de Cálcio Tipo T/genética , Ciclopropanos/farmacologia , Ciclopropanos/uso terapêutico , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/genética , Inflamação/genética , Inflamação/metabolismo , Mibefradil/farmacologia , Mibefradil/uso terapêutico , Camundongos , Naftalenos/farmacologia , Naftalenos/uso terapêutico , Neuralgia/tratamento farmacológico , Neuralgia/genética , Neurônios/efeitos dos fármacos
5.
PLoS One ; 9(4): e91467, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24705276

RESUMO

We recently showed that streptozotocin (STZ) injections in rats lead to the development of painful peripheral diabetic neuropathy (PDN) accompanied by enhancement of CaV3.2 T-type calcium currents (T-currents) and hyperexcitability in dorsal root ganglion (DRG) neurons. Here we used the classical peripherally acting T-channel blocker mibefradil to examine the role of CaV3.2 T-channels as pharmacological targets for treatment of painful PDN. When administered intraperitoneally (i.p.), at clinically relevant doses, mibefradil effectively alleviated heat, cold and mechanical hypersensitivities in STZ-treated diabetic rats in a dose-dependent manner. We also found that CaV3.2 antisense (AS)-treated diabetic rats exhibit a significant decrease in painful PDN compared with mismatch antisense (MIS)-treated diabetic rats. Co-treatment with mibefradil (9 mg/kg i.p.) resulted in reversal of heat, cold and mechanical hypersensitivity in MIS-treated but not in AS-treated diabetic rats, suggesting that mibefradil and CaV3.2 AS share the same cellular target. Using patch-clamp recordings from acutely dissociated DRG neurons, we demonstrated that mibefradil similarly blocked T-currents in diabetic and healthy rats in a voltage-dependent manner by stabilizing inactive states of T-channels. We conclude that antihyperalgesic and antiallodynic effects of mibefradil in PDN are at least partly mediated by inhibition of CaV3.2 channels in peripheral nociceptors. Hence, peripherally acting voltage-dependent T-channel blockers could be very useful in the treatment of painful symptoms of PDN.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/fisiologia , Neuropatias Diabéticas/tratamento farmacológico , Hiperalgesia/prevenção & controle , Mibefradil/uso terapêutico , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Neuropatias Diabéticas/patologia , Feminino , Mibefradil/farmacologia , Ratos , Ratos Sprague-Dawley , Estreptozocina
6.
Clinics (Sao Paulo) ; 69(1): 61-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24473561

RESUMO

OBJECTIVE: Numerous recent studies suggest that abnormal intracellular calcium concentration ([Ca2+]i) is a common defect in diabetic animal models and patients. Abnormal calcium handling is an important mechanism in the defective pancreatic ß-cell function in type 2 diabetes. T-type Ca2+ channel antagonists lower blood glucose in type 2 diabetes, but the mechanism remains unknown. METHODS: We examined the effect of the Ca2+ channel antagonist mibefradil on blood glucose in male db/db mice and phenotypically normal heterozygous mice by intraperitoneal injection. RESULTS: Mibefradil (15 mg/kg, i.p., b.i.d.) caused a profound reduction of fasting blood glucose from 430.92±20.46 mg/dl to 285.20±5.74 mg/dl in three days. The hypoglycemic effect of mibefradil was reproduced by NNC 55-0396, a compound structurally similar to mibefradil but more selective for T-type Ca2+ channels, but not by the specific L-type Ca2+ channel blocker nicardipine. Mibefradil did not show such hypoglycemic effects in heterozygous animals. In addition, triglycerides, basal insulin and food intake were significantly decreased by mibefradil treatment in the db/db mice but not in the controls. Western blot analysis, immunohistochemistry and immunofluorescence staining showed a significantly increased expression of T-type Ca2+ channel α-subunits Cav3.1 and Cav3.2 in liver and brain tissues from db/db mice compared to those from heterozygous animals. CONCLUSIONS: Collectively, these results suggest that T-type Ca2+ channels are potential therapeutic targets for antidiabetic drugs.


Assuntos
Glicemia/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Mibefradil/uso terapêutico , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/administração & dosagem , Modelos Animais de Doenças , Ingestão de Alimentos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Imuno-Histoquímica , Injeções Intraperitoneais , Fígado/efeitos dos fármacos , Masculino , Ilustração Médica , Mibefradil/administração & dosagem , Camundongos , Reprodutibilidade dos Testes , Fatores de Tempo , Resultado do Tratamento
7.
Clinics ; 69(1): 61-67, 1/2014. graf
Artigo em Inglês | LILACS | ID: lil-697715

RESUMO

OBJECTIVE: Numerous recent studies suggest that abnormal intracellular calcium concentration ([Ca2+]i) is a common defect in diabetic animal models and patients. Abnormal calcium handling is an important mechanism in the defective pancreatic β-cell function in type 2 diabetes. T-type Ca2+ channel antagonists lower blood glucose in type 2 diabetes, but the mechanism remains unknown. METHODS: We examined the effect of the Ca2+ channel antagonist mibefradil on blood glucose in male db/db mice and phenotypically normal heterozygous mice by intraperitoneal injection. RESULTS: Mibefradil (15 mg/kg, i.p., b.i.d.) caused a profound reduction of fasting blood glucose from 430.92±20.46 mg/dl to 285.20±5.74 mg/dl in three days. The hypoglycemic effect of mibefradil was reproduced by NNC 55-0396, a compound structurally similar to mibefradil but more selective for T-type Ca2+ channels, but not by the specific L-type Ca2+ channel blocker nicardipine. Mibefradil did not show such hypoglycemic effects in heterozygous animals. In addition, triglycerides, basal insulin and food intake were significantly decreased by mibefradil treatment in the db/db mice but not in the controls. Western blot analysis, immunohistochemistry and immunofluorescence staining showed a significantly increased expression of T-type Ca2+ channel α-subunits Cav3.1 and Cav3.2 in liver and brain tissues from db/db mice compared to those from heterozygous animals. CONCLUSIONS: Collectively, these results suggest that T-type Ca2+ channels are potential therapeutic targets for antidiabetic drugs. .


Assuntos
Animais , Masculino , Camundongos , Glicemia/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Mibefradil/uso terapêutico , Western Blotting , Encéfalo/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/administração & dosagem , Modelos Animais de Doenças , Ingestão de Alimentos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Imuno-Histoquímica , Injeções Intraperitoneais , Fígado/efeitos dos fármacos , Ilustração Médica , Mibefradil/administração & dosagem , Reprodutibilidade dos Testes , Fatores de Tempo , Resultado do Tratamento
8.
J Neurooncol ; 111(2): 97-102, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23086436

RESUMO

Glioblastoma multiforme (GBM) is a devastating disease with a dismal prognosis and a very limited response to treatment. The current standard of care for GBM usually consists of surgery, radiation and chemotherapy with the alkylating agent temozolomide, although resistance to this drug is common. The predominant mechanism of action of temozolomide is methylation of guanine residues although this can be reversed by methylguanine methyltransferase (MGMT) as well as other DNA repair systems. The presence of methylguanine causes abortive DNA synthesis with subsequent apoptosis. This suggests that the closer a particular cell is to S phase when it is exposed to temozolomide the more likely it is to die since repair enzymes will have had less time to reverse the damage. T type calcium channel inhibitors can stop the entry of extracellular calcium that is necessary for transit past the G1/S boundary. As a result, T type calcium channel blockers can slow the growth of cancer cells, but do not generally kill them. Though slowing the growth of cancer cells is important in its own right, it also provides a therapeutic strategy in which a T type channel blocker is administered then withdrawn followed by the administration of temozolomide. We show here that imposing this cell cycle restriction increases the efficacy of subsequently administered temozolomide in immunodeficient mice bearing various human GBM xenograft lines. We also present data that MGMT expressing GBM tumors, which are temozolomide resistant, may be rendered more sensitive by this strategy.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Mibefradil/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/etiologia , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Modelos Animais de Doenças , Esquema de Medicação , Quimioterapia Combinada , Humanos , Estimativa de Kaplan-Meier , Camundongos , Transplante de Neoplasias/métodos , Neoplasias Cutâneas/etiologia , Temozolomida , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Neurosurg ; 118(4): 830-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23198803

RESUMO

OBJECT: The survival of patients with high-grade gliomas remains unfavorable. Mibefradil, a T-type calcium channel inhibitor capable of synchronizing dividing cells at the G1 phase, has demonstrated potential benefit in conjunction with chemotherapeutic agents for gliomas in in vitro studies. In vivo study of mibefradil and radiosurgery is lacking. The authors used an intracranial C6 glioma model in rats to study tumor response to mibefradil and radiosurgery. METHODS: Two weeks after implantation of C6 cells into the animals, each rat underwent MRI every 2 weeks thereafter for 8 weeks. After tumor was confirmed on MRI, the rats were randomly assigned to one of the experimental groups. Tumor volumes were measured on MR images. Experimental Group 1 received 30 mg/kg of mibefradil intraperitoneally 3 times a day for 1 week starting on postoperative day (POD) 15; Group 2 received 8 Gy of cranial radiation via radiosurgery delivered on POD 15; Group 3 underwent radiosurgery on POD 15, followed by 1 week of mibefradil; and Group 4 received mibefradil on POD 15 for 1 week, followed by radiosurgery sometime from POD 15 to POD 22. Twenty-seven glioma-bearing rats were analyzed. Survival was compared between groups using Kaplan-Meier methodology. RESULTS: Median survival in Groups 1, 2, 3, and 4 was 35, 31, 43, and 52 days, respectively (p = 0.036, log-rank test). Two animals in Group 4 survived to POD 60, which is twice the expected survival of untreated animals in this model. Analysis of variance and a post hoc test indicated no tumor volume differences on PODs 15 and 29. However, significant volume differences were found on POD 43; mean tumor volumes for Groups 1, 2, 3, and 4 were 250, 266, 167, and 34 mm(3), respectively (p = 0.046, ANOVA). A Cox proportional hazards regression test showed survival was associated with tumor volume on POD 29 (p = 0.001) rather than on POD 15 (p = 0.162). In vitro assays demonstrated an appreciable and dose-dependent increase in apoptosis between 2- and 7-µM concentrations of mibefradil. CONCLUSIONS: Mibefradil response is schedule dependent and enhances survival and reduces glioblastoma when combined with ionizing radiation.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/cirurgia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/cirurgia , Mibefradil/uso terapêutico , Radiocirurgia , Animais , Neoplasias Encefálicas/mortalidade , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glioblastoma/mortalidade , Imageamento por Ressonância Magnética , Modelos de Riscos Proporcionais , Ratos , Ratos Sprague-Dawley , Estudos Retrospectivos , Taxa de Sobrevida , Resultado do Tratamento
10.
Eur J Pharmacol ; 659(1): 30-6, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21256842

RESUMO

NNC 55-0396 [(1S,2S)-2-(2-(N-[(3-benzimidazol-2-yl)propyl]-N-methylamino)ethyl)-6-fluoro-1,2, 3,4-tetrahydro-1-isopropyl-2-naphtyl cyclopropanecarboxylate dihydrochloride], is a mibefradil derivative that retains potent in vitro T-type calcium channel antagonist efficacy. We compared the two compounds for behavioral toxicity, effects on cytochrome P450 activity, and efficacy against tremor in the γ-aminobutyric acid type A (GABAA) receptor subunit α1-null mouse, and the harmaline tremor model of essential tremor in wild-type mice. NNC 55-0396 was better tolerated than mibefradil in the horizontal wire test of sedation/motor function, with 3/6 failing at 300 and 30mg/kg respectively. To assess for a potential interaction with harmaline, mice were given the drugs, followed by harmaline or vehicle, and tested 30min later in the inverted wire grid test. Mibefradil exacerbated, whereas NNC 55-0396 ameliorated harmaline-induced test deficits. In mouse liver microsomes, NNC 55-0396 was a less potent inhibitor of harmaline O-demethylation than mibefradil (Ki: 0.95 and 0.29µM respectively), and also less potent at inhibiting testosterone 6-ß-hydroxylation (Ki: 0.71 and 0.12µM respectively). In the GABAA α1-null model, NNC 55-0396 but not mibefradil, (each at 20mg/kg), suppressed tremor while NNC 55-0396 at 12.5mg/kg suppressed harmaline-induced tremor by half by 20-100min, whereas mibefradil at the same dose did not significantly affect tremor. In contrast to mibefradil, NNC 55-0396 is well tolerated and suppresses tremor, and exerts less cytochrome P450 inhibition. These results suggest potential clinical utility for NNC 55-0396 or similar derivatives as a T-type calcium antagonist.


Assuntos
Comportamento Animal/efeitos dos fármacos , Benzimidazóis/química , Benzimidazóis/farmacologia , Ciclopropanos/química , Ciclopropanos/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Tremor Essencial/tratamento farmacológico , Mibefradil/química , Mibefradil/farmacologia , Naftalenos/química , Naftalenos/farmacologia , Animais , Benzimidazóis/uso terapêutico , Ciclopropanos/uso terapêutico , Modelos Animais de Doenças , Tremor Essencial/enzimologia , Tremor Essencial/metabolismo , Deleção de Genes , Harmalina/metabolismo , Hidroxilação/efeitos dos fármacos , Metilação/efeitos dos fármacos , Mibefradil/uso terapêutico , Camundongos , Naftalenos/uso terapêutico , Receptores de GABA-A/deficiência , Receptores de GABA-A/genética , Relação Estrutura-Atividade , Testosterona/metabolismo
11.
Fundam Clin Pharmacol ; 25(4): 469-78, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21039820

RESUMO

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.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Ataque Isquêmico Transitório/tratamento farmacológico , Mibefradil/uso terapêutico , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Pimozida/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Região CA1 Hipocampal/patologia , Bloqueadores dos Canais de Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacocinética , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/metabolismo , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Glucose/deficiência , Células HEK293 , Humanos , Hipóxia , Ataque Isquêmico Transitório/sangue , Ataque Isquêmico Transitório/mortalidade , Ataque Isquêmico Transitório/patologia , Ácido Láctico/sangue , Masculino , Potenciais da Membrana/fisiologia , Mibefradil/metabolismo , Mibefradil/farmacocinética , Mibefradil/farmacologia , Neurônios/patologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/farmacologia , Pimozida/farmacologia , Ratos , Ratos Sprague-Dawley , Análise de Sobrevida , Técnicas de Cultura de Tecidos , Transfecção
12.
Pain ; 150(1): 183-191, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20546998

RESUMO

Hydrogen sulfide (H(2)S) formed from l-cysteine by multiple enzymes including cystathionine-gamma-lyase (CSE) is now considered a gasotransmitter in the mammalian body. Our previous studies have shown that H(2)S activates/sensitizes Ca(v)3.2 T-type Ca(2+) channels, leading to facilitation of somatic and visceral nociception, and that CSE-derived endogenous H(2)S participates in inflammatory pain. Here, we show novel evidence for involvement of the endogenous H(2)S-Ca(v)3.2 pathway in neuropathic pain. In the rat subjected to the right L5 spinal nerve cutting (L5SNC), a neuropathic pain model, i.p. administration of dl-propargylglycine (PPG) and beta-cyanoalanine, irreversible and reversible CSE inhibitors, respectively, strongly suppressed the neuropathic hyperalgesia/allodynia. The anti-hyperalgesic effect of PPG was reversed by intraplantar administration of NaHS, a donor for H(2)S, in the L5SNC rat. Intraplantar administration or topical application of mibefradil, a T-type Ca(2+) channel blocker, reversed hyperalgesia in the L5SNC rat. The protein levels of Ca(v)3.2, but not CSE, in the ipsilateral L4, L5 and L6 dorsal root ganglia were dramatically upregulated in the L5SNC rat. Finally, silencing of Ca(v)3.2 in DRG by repeated intrathecal administration of Ca(v)3.2-targeting siRNA significantly attenuated the neuropathic hyperalgesia in the L5SNC rat. In conclusion, our data suggest that Ca(v)3.2 T-type Ca(2+) channels in sensory neurons are upregulated and activated/sensitized by CSE-derived endogenous H(2)S after spinal nerve injury, contributing to the maintenance of neuropathic pain. We thus propose that Ca(v)3.2 and CSE could be targets for the development of therapeutic drugs for the treatment of neuropathic pain.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Sulfeto de Hidrogênio/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Regulação para Cima/fisiologia , Animais , Western Blotting , Bloqueadores dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/uso terapêutico , Hiperalgesia/tratamento farmacológico , Imuno-Histoquímica , Masculino , Mibefradil/farmacologia , Mibefradil/uso terapêutico , Neuralgia/tratamento farmacológico , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Limiar da Dor/fisiologia , RNA Interferente Pequeno , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nervos Espinhais/lesões
13.
Pharmacology ; 85(5): 295-300, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20453553

RESUMO

This study aimed to elucidate the role of T-type calcium channels in the nociceptive signal transmission at the spinal level. The chronic compression of dorsal root ganglion (CCD) rat model was adopted. Three doses (50, 100 and 200 microg in groups Mib50, Mib100 and Mib200, respectively) of specific T-type Ca2+ channel inhibitors mibefradil (Mib) or normal saline (NS) were intrathecally administered on the 5th day after the CCD model had been established. The paw withdrawal latency from a noxious thermal stimulus and paw withdrawal mechanical threshold of von Frey filament was used to measure the thermal hyperalgesia and tactile allodynia, respectively. Lumbar spinal cords of the rats isolated on the 5th day after the operation were prepared to measure the mRNA expression of T-type (Cav3.1, Cav3.2 and Cav3.3) calcium channel with RT-PCR methods. The results demonstrated that CCD rats produced reliable thermal hyperalgesia and tactile allodynia after surgery. The intrathecal administration of Mib significantly suppressed thermal hyperalgesia and allodynia in CCD rats (p< 0.01), and the inhibitory effect lasted for 2 h. However, only Cav3.2 and Cav3.3 T-type calcium channel mRNA were detected in the lumbar spinal cord of rats, and there were no Cav3.1 calcium channels. Compared with native and sham groups, the Cav3.2 and Cav3.3 calcium channel mRNA expression increased significantly (p < 0.05). These data support the view that spinal T-type calcium (Cav3.2 and Cav3.3 but not Cav3.1) channels may play an important role in the pathogenesis of neuropathic pain.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Gânglios Espinais/lesões , Síndromes de Compressão Nervosa/fisiopatologia , Neuralgia/fisiopatologia , Animais , Comportamento Animal/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/administração & dosagem , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/genética , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Temperatura Alta , Hiperalgesia/tratamento farmacológico , Vértebras Lombares/metabolismo , Masculino , Mibefradil/administração & dosagem , Mibefradil/uso terapêutico , Síndromes de Compressão Nervosa/metabolismo , Neuralgia/tratamento farmacológico , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Fatores de Tempo , Tato
15.
Pain ; 142(1-2): 127-32, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19167819

RESUMO

Hydrogen sulfide (H2S), a gasotransmitter, facilitates membrane currents through T-type Ca2+ channels, and intraplantar (i.pl.) administration of NaHS, a donor of H2S, causes prompt hyperalgesia in rats. In this context, we asked whether intrathecal (i.t.) administration of NaHS could mimic the hyperalgesic effect of i.pl. NaHS in rats, and then examined if Cav3.2 isoform of T-type Ca2+ channels contributed to the pro-nociceptive effects of i.t. and i.pl. NaHS. Either i.t. or i.pl. administration of NaHS rapidly decreased nociceptive threshold in rats, as determined by the paw pressure method. The hyperalgesia caused by i.t. and i.pl. NaHS was abolished by co-administration of mibefradil, a pan-T-type Ca2+ channel inhibitor, and also suppressed by pretreatment with i.t. and i.pl. zinc chloride, known to preferentially inhibit Cav3.2 among T-type Ca2+ channel isoforms, respectively. Repeated i.t. administration of antisense oligodeoxynucleotides (ODNs) targeting rat Cav3.2, but not mismatch ODNs, caused silencing of Cav3.2 protein in the dorsal root ganglia and spinal cord, and then attenuated the hyperalgesia induced by either i.t. or i.pl. NaHS. Our findings thus establish that spinal and peripheral NaHS/H2S activates or sensitizes Cav3.2 T-type Ca2+ channels expressed in the primary afferents and/or spinal nociceptive neurons, leading to sensitization of nociceptive processing and hyperalgesia.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/metabolismo , Sulfeto de Hidrogênio/efeitos adversos , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Mibefradil/uso terapêutico , Limiar da Dor/fisiologia , Medula Espinal/efeitos dos fármacos , Análise de Variância , Animais , Canais de Cálcio Tipo T/genética , Cerebelo/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Wistar
16.
J Pharmacol Exp Ther ; 325(3): 723-31, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18326812

RESUMO

Among the L-type calcium channel blockers (CCBs), particularly dihydropyridines like nifedipine [1,4-dihydro-2,6-dimethyl-4-(2-nitrophenyl)-3,5-pyridinedicarboxylic acid dimethyl ester], a common adverse effect is vasodilatory edema. Newer CCBs, such as the T- and L-type CCB, mibefradil [(1S,2S)-2-[2[[3-(2-benzimidazolylpropyl]methylamino]ethyl]-6-fluoro-1,2,3,4-tetrahydro-1-isopropyl-2-naphthyl methoxyacetate dihydrochloride hydrate], demonstrate antihypertensive efficacy similar to that of their predecessors but seem to have a reduced propensity to cause edema. Using a magnetic resonance imaging (MRI) T(2) mapping technique, we investigated the ability of mibefradil to reduce extracellular water accumulation caused by the L-type CCB, nifedipine, in the hindleg skeletal muscle of the spontaneously hypertensive rat. Mibefradil (10 mg/kg i.v.) and nifedipine (1 mg/kg i.v.) lowered mean arterial blood pressure by 97 +/- 5 and 77 +/- 4 mm Hg, respectively. MRI edema index (expressed as percentage increase of integral T(2) over predrug control) was significantly higher with nifedipine (2606 +/- 86%; p < 0.05) than with mibefradil (981 +/- 171%) measured 30 to 60 min after the start of drug infusion. The hindleg edema caused by nifedipine was dose dependently decreased by coadministration of mibefradil (0, 0.3, or 3 mg/kg). The hindleg edema formation was not due to albumin leakage into the interstitial space based on immunostaining. However, a 4.2-fold increase in the arterial L-/T-type CC mRNA expression ratio was observed compared with the venous L/T ratio as shown by quantitative reverse transcription polymerase chain reaction. These results demonstrate the novel utility of MRI to measure extravascular water after acute exposure to CCBs and indicate that T-type CCB activity may reduce L-type CCB-induced vasodilatory edema in the skeletal muscle vasculature, possibly by a differential effect on arteriole and venule dilatation.


Assuntos
Anti-Hipertensivos/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo T/fisiologia , Edema/induzido quimicamente , Edema/tratamento farmacológico , Hipertensão/tratamento farmacológico , Mibefradil/uso terapêutico , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo T/genética , Edema/patologia , Edema/fisiopatologia , Artéria Femoral/metabolismo , Membro Posterior , Hipertensão/patologia , Hipertensão/fisiopatologia , Imageamento por Ressonância Magnética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Nifedipino/uso terapêutico , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley
17.
J Hypertens ; 25(1): 169-75, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17143189

RESUMO

OBJECTIVES: Calcium antagonists are vasodilating drugs, which may cause reflex activation of the sympathetic nervous system with potentially untoward effects. We studied the effects of long-term treatment with amlodipine, a long-acting dihydropyridine-type calcium antagonist, and mibefradil, a phenylalkylamine-type calcium antagonist, on sympathetic nerve activity. METHODS: Fourteen patients with primary hypertension participated in a double-blind, cross-over study comparing the effects of 6 weeks of treatment with mibefradil 100 mg daily and amlodipine 10 mg daily. Heart rate, direct arterial blood pressure and cardiac output by echocardiography were registered. Global sympathetic activity was estimated using a [3H]noradrenaline isotope dilution method with arterial and venous sampling; cardiac sympathetic activity was assessed indirectly by heart rate variability and tissue velocity echocardiography. RESULTS: Both drugs lowered mean arterial pressure; the decrease was more pronounced with mibefradil (from 118 +/- 3 to 99 +/- 2 mmHg, compared to 118 +/- 3 to 104 +/- 2 mmHg for amlodipine, P < 0.01 between drugs). Mibefradil decreased heart rate (66 +/- 2 to 57 +/- 2 bpm), whereas amlodipine caused a slight increase (66 +/- 2 to 70 +/- 2 bpm; P < 0.001 between drugs) and tended to increase cardiac output. Noradrenaline spillover increased similarly with the two drugs, from 3.44 +/- 0.27 to 5.20 +/- 0.48 nmol/min per m2(P < 0.01) during mibefradil and to 5.72 +/- 0.49 nmol/min per m2 (P < 0.001) during amlodipine. There were minor effects on cardiac sympatho-vagal balance, but systolic and diastolic myocardial velocities were increased similarly by both drugs. CONCLUSIONS: Mibefradil and amlodipine treatment increase global sympathetic nerve activity similarly during long-term treatment, despite opposite effects on heart rate. Increases in myocardial velocities suggest concomitant cardiac sympathetic activation.


Assuntos
Anlodipino/uso terapêutico , Anti-Hipertensivos/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Hipertensão/tratamento farmacológico , Mibefradil/uso terapêutico , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Anlodipino/farmacologia , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Débito Cardíaco/efeitos dos fármacos , Estudos Cross-Over , Método Duplo-Cego , Feminino , Coração/inervação , Frequência Cardíaca/efeitos dos fármacos , Humanos , Hipertensão/sangue , Hipertensão/diagnóstico por imagem , Hipertensão/fisiopatologia , Masculino , Mibefradil/farmacologia , Pessoa de Meia-Idade , Norepinefrina/sangue , Fatores de Tempo , Ultrassonografia
18.
Dig Dis Sci ; 51(8): 1454-60, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16868826

RESUMO

The purpose of the present study was to investigate whether mibefradil can reduce oxidative stress and histologic damage in the rat small bowel subjected to mesenteric ischemia and reperfusion injury. Thirty Sprague-Dawley rats weighing between 210 and 220 g were divided into three groups, each containing 10 rats: group 1, sham operation; group 2, untreated ischemia-reperfusion; and group 3, ischemia-reperfusion plus mibefradil treatment group. Intestinal ischemia for 45 min and reperfusion for 60 min were applied. Ileal specimens were obtained to determine the tissue levels of MDA, CAT, SOD, and GSH-Px and histologic changes. In group 2, MDA values were significantly increased compared to those in groups 1 and 3. In addition, SOD, CAT, and GSH-Px values decreased significantly in group 2 compared to groups 1 and 3. The intestinal injury score increased significantly in group 2 and 3 rats compared to group 1 rats. However, this increase was reduced in group 3 rats compared to group 2. Histopathologically, the rats in group 1 had essentially normal testicular architecture. In group 2 rats, the lesions varied between grade 3 and grade 5. In contrast, most of the specimens in the mibefradil-treated group 3 showed grade 1 injury. Mibefradil plays a role in attenuating reperfusion injury of the small intestine by depressing free radical production and mucosal injury score and regulating postischemic intestinal perfusion while restoring intestinal microcirculatory blood flow and encountered histologic injury.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/efeitos dos fármacos , Mucosa Intestinal/patologia , Mesentério/irrigação sanguínea , Mibefradil/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Índice de Gravidade de Doença , Superóxido Dismutase/metabolismo , Resultado do Tratamento
19.
J Am Coll Cardiol ; 44(1): 57-62, 2004 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-15234407

RESUMO

OBJECTIVES: The aim of the study was to assess the angiographic and clinical benefits of the calcium T-channel blocker, mibefradil, in the coronary slow flow phenomenon (CSFP). BACKGROUND: The CSFP is characterized by delayed vessel opacification on angiography (Thrombolysis In Myocardial Infarction [TIMI]-2 flow) in the absence of obstructive epicardial coronary disease and is often associated with recurrent chest pain. METHODS: A total of 10 CSFP patients (46 +/- 9 years) underwent angiography before and 30 min after 50 mg mibefradil; off-line blinded analysis of angiographic data included comparisons of epicardial vessel diameter, TIMI flow grade and TIMI frame count. We also performed a randomized, double-blind, placebo-controlled, cross-over study to examine the long-term efficacy of mibefradil 100 mg/day on the frequency of total angina, prolonged angina (i.e., persisting >20 min) episodes, and sublingual nitrate consumption, during consecutive one-month treatment periods in 20 patients (age 51 +/- 12 years) with the CSFP. RESULTS: Without changing epicardial vessel diameter or rate-pressure product, mibefradil reduced the number of vessels exhibiting TIMI-2 flow from 18 to 5; furthermore, mibefradil significantly improved the TIMI frame count only in those vessels exhibiting TIMI-2 flow (28 +/- 18%, p < 0.005). Compared with placebo, mibefradil significantly reduced total angina frequency by 56% (p < 0.001), prolonged episodes of angina by 74% (p < 0.001), and sublingual nitrate consumption by 59% (p < 0.01); furthermore, mibefradil improved physical quality of life as assessed by the Health Outcome Study Short Form-36. CONCLUSIONS: These angiographic and clinical improvements produced by mibefradil support a microspastic pathogenesis of the CSFP.


Assuntos
Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Angiografia Coronária , Doença das Coronárias/tratamento farmacológico , Doença das Coronárias/fisiopatologia , Mibefradil/uso terapêutico , Adulto , Biomarcadores/sangue , Pressão Sanguínea/efeitos dos fármacos , Proteína C-Reativa/metabolismo , Bloqueadores dos Canais de Cálcio/administração & dosagem , Doença das Coronárias/diagnóstico , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/fisiopatologia , Estudos Cross-Over , Diástole/efeitos dos fármacos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Ecocardiografia , Eletrocardiografia Ambulatorial , Feminino , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Mibefradil/administração & dosagem , Pessoa de Meia-Idade , Cooperação do Paciente , Estatística como Assunto , Sístole/efeitos dos fármacos , Resultado do Tratamento , Troponina I/sangue
20.
J Cardiovasc Pharmacol ; 42(6): 697-702, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14639089

RESUMO

Although nifedipine and other conventional calcium antagonists elicit preferential vasodilation of renal afferent arterioles, we demonstrate that mibefradil and nickel, T-type calcium channel blockers, reverse the angiotensin II-induced constriction of both afferent and efferent arterioles. Since the angiotensin II-induced vasoconstriction involves inositol trisphosphate (IP3)-induced calcium release from the sarcoplasmic reticulum in the afferent arteriole, and both IP3- and protein kinase C (PKC)-mediated pathways in the efferent arteriole, we investigated the cellular mechanism for the mibefradil-induced dilation of angiotensin II-constricted renal arterioles, using the isolated perfused hydronephrotic rat kidney. Mibefradil caused a dose-dependent dilation of angiotensin II-constricted afferent and efferent arterioles, with 88 +/- 9% and 74 +/- 10% reversal observed at 1 micromol/L, respectively. The blockade of PKC by staurosporine did not alter the mibefradil-induced vasodilator responses of either arterioles (P > 0.5). In contrast, the pretreatment with thapsigargin, which predominantly blocked the IP3-mediated intracellular calcium release, prevented the afferent arteriolar constrictor response to angiotensin II, but caused a significant constriction of efferent arterioles. The subsequent addition of mibefradil had no effect on the efferent arteriolar diameter. Furthermore, the efferent arteriolar constriction induced by direct PKC activation by phorbol myristate acetate was refractory to mibefradil, but completely reversed by LOE908, a nonselective cation channel blocker. In summary, mibefradil markedly dilates the angiotensin II-induced renal arteriolar constriction; the action of mibefradil is most likely mediated by the inhibition of the IP3-mediated pathway, but the inhibitory action on the PKC pathway appears modest.


Assuntos
Angiotensina II/antagonistas & inibidores , Bloqueadores dos Canais de Cálcio/uso terapêutico , Hidronefrose/tratamento farmacológico , Mibefradil/uso terapêutico , Circulação Renal/efeitos dos fármacos , Vasoconstritores/antagonistas & inibidores , Vasodilatadores/uso terapêutico , Análise de Variância , Animais , Masculino , Ratos , Ratos Endogâmicos WKY
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...