Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacol Sci ; 152(2): 86-89, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37169483

RESUMO

Cav3.2, a T-type calcium channel (T-channel) family member, is expressed in the nociceptors and spinal cord, and its activity is largely suppressed by zinc under physiological conditions. In rats, intrathecal and intraplantar administration of a zinc chelator, TPEN, caused T-channel-dependent mechanical hyperalgesia, and the intraplantar, but not intrathecal, TPEN induced Cav3.2 upregulation in the dorsal root ganglion. In mice, intraplantar TPEN also caused mechanical allodynia, which was abolished by T-channel inhibitors or Cav3.2 gene deletion. Together, spinal and peripheral zinc deficiency appears to enhance Cav3.2 activity in the spinal postsynaptic neurons and nociceptors, respectively, thereby promoting pain.


Assuntos
Canais de Cálcio Tipo T , Hiperalgesia , Ratos , Camundongos , Animais , Hiperalgesia/induzido quimicamente , Roedores , Quelantes , Zinco , Canais de Cálcio Tipo T/genética , Gânglios Espinais
2.
J Pharmacol Sci ; 140(3): 310-312, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31492577

RESUMO

We tested whether genetic deletion of Cav3.2 T-type Ca2+ channels abolishes hydrogen sulfide (H2S)-mediated pain signals in mice. In Cav3.2-expressing HEK293 cells, Na2S, an H2S donor, at 100 µM clearly increased Ba2+ currents, as assessed by whole-cell patch-clamp recordings. In wild-type C57BL/6 mice, intraplantar and intracolonic administration of Na2S evoked mechanical allodynia and visceral nociceptive behavior, respectively, which were abolished by TTA-A2, a T-type Ca2+ channel blocker. In Cav3.2-knockout mice of a C57BL/6 background, Na2S caused neither somatic allodynia nor colonic nociception. Our study thus provides definitive evidence for an essential role of Cav3.2 in H2S-dependent somatic and colonic pain.


Assuntos
Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/metabolismo , Sulfeto de Hidrogênio/farmacologia , Dor Nociceptiva/metabolismo , Transdução de Sinais/efeitos dos fármacos , Dor Visceral/metabolismo , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Wistar , Canal de Cátion TRPA1/metabolismo
3.
J Pharmacol Sci ; 133(1): 57-60, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28081948

RESUMO

Given that Cav3.2 T-type Ca2+ channels were functionally regulated by asparagine (N)-linked glycosylation, we examined effects of high glucose on the function of Cav3.2, known to regulate secretory function, in neuroendocrine-like differentiated prostate cancer LNCaP cells. High glucose accelerated the increased channel function and overexpression of Cav3.2 during neuroendocrine differentiation, the former prevented by enzymatic inhibition of N-glycosylation and cleavage of N-glycans. Hyperglycemia thus appears to induce N-linked glycosylation-mediated functional upregulation and overexpression of Cav3.2 in neuroendocrine-like differentiated prostate cancer cells.


Assuntos
Canais de Cálcio Tipo T/biossíntese , Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Glucose/farmacologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Regulação para Cima/efeitos dos fármacos , 1-Metil-3-Isobutilxantina/farmacologia , Bucladesina/farmacologia , Linhagem Celular Tumoral , Glicosilação/efeitos dos fármacos , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Tunicamicina/farmacologia
4.
Redox Biol ; 59: 102579, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36563535

RESUMO

Poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132), an organogermanium, is hydrolyzed to 3-(trihydroxygermyl)propanoic acid (THGP) in aqueous solutions, and reduces inflammation, pain and cancer, whereas the underlying mechanisms remain unknown. Sulfides including H2S, a gasotransmitter, generated from l-cysteine by some enzymes including cystathionine-γ-lyase (CSE), are pro-nociceptive, since they enhance Cav3.2 T-type Ca2+ channel activity expressed in the primary afferents, most probably by canceling the channel inhibition by Zn2+ linked via coordinate bonding to His191 of Cav3.2. Given that germanium is reactive to sulfur, we tested whether THGP would directly trap sulfide, and inhibit sulfide-induced enhancement of Cav3.2 activity and sulfide-dependent pain in mice. Using mass spectrometry and 1H NMR techniques, we demonstrated that THGP directly reacted with sulfides including Na2S and NaSH, and formed a sulfur-containing reaction product, which decreased in the presence of ZnCl2. In Cav3.2-transfected HEK293 cells, THGP inhibited the sulfide-induced enhancement of T-type Ca2+ channel-dependent membrane currents. In mice, THGP, administered systemically or locally, inhibited the mechanical allodynia caused by intraplantar Na2S. In the mice with cyclophosphamide-induced cystitis and cerulein-induced pancreatitis, which exhibited upregulation of CSE in the bladder and pancreas, respectively, systemic administration of THGP as well as a selective T-type Ca2+ channel inhibitor suppressed the cystitis-related and pancreatitis-related visceral pain. These data suggest that THGP traps sulfide and inhibits sulfide-induced enhancement of Cav3.2 activity, leading to suppression of Cav3.2-dependent pain caused by sulfide applied exogenously and generated endogenously.


Assuntos
Canais de Cálcio Tipo T , Cistite , Sulfeto de Hidrogênio , Pancreatite , Dor Visceral , Camundongos , Humanos , Animais , Células HEK293 , Canais de Cálcio Tipo T/fisiologia , Sulfetos/farmacologia , Cistite/induzido quimicamente , Sulfeto de Hidrogênio/metabolismo
5.
Eur J Pharmacol ; 887: 173576, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32949597

RESUMO

Given the role of Cav3.2 isoform among T-type Ca2+ channels (T-channels) in somatic and visceral nociceptive processing, we analyzed the contribution of Cav3.2 to butyrate-induced colonic pain and nociceptor hypersensitivity in mice, to evaluate whether Cav3.2 could serve as a target for treatment of visceral pain in irritable bowel syndrome (IBS) patients. Mice of ddY strain, and wild-type and Cav3.2-knockout mice of a C57BL/6J background received intracolonic administration of butyrate twice a day for 3 days. Referred hyperalgesia in the lower abdomen was assessed by von Frey test, and colonic hypersensitivity to distension by a volume load or chemicals was evaluated by counting nociceptive behaviors. Spinal phosphorylated ERK was detected by immunohistochemistry. Cav3.2 knockdown was accomplished by intrathecal injection of antisense oligodeoxynucleotides. Butyrate treatment caused referred hyperalgesia and colonic hypersensitivity to distension in ddY mice, which was abolished by T-channel blockers and/or Cav3.2 knockdown. Butyrate also increased the number of spinal phosphorylated ERK-positive neurons following colonic distension in the anesthetized ddY mice. The butyrate-treated ddY mice also exhibited T-channel-dependent colonic hypersensitivity to intracolonic Na2S, known to enhance Cav3.2 activity, and TRPV1, TRPA1 or proteinase-activated receptor 2 (PAR2) agonists. Wild-type, but not Cav3.2-knockout, mice of a C57BL/6J background, after treated with butyrate, mimicked the T-channel-dependent referred hyperalgesia and colonic hypersensitivity in butyrate-treated ddY mice. Our study provides definitive evidence for an essential role of Cav3.2 in the butyrate-induced colonic pain and nociceptor hypersensitivity, which might serve as a target for treatment of visceral pain in IBS patients.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Doenças do Colo/induzido quimicamente , Nociceptores/efeitos dos fármacos , Dor Visceral/induzido quimicamente , Animais , Butiratos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/genética , Doenças do Colo/genética , Sulfeto de Hidrogênio/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/fisiopatologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medição da Dor/efeitos dos fármacos , Dor Visceral/genética
6.
Eur J Pharmacol ; 888: 173587, 2020 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-32971090

RESUMO

Overexpression of Cav3.2 T-type Ca2+ channels in L4 dorsal root ganglion (DRG) participates in neuropathic pain after L5 spinal nerve cutting (L5SNC) in rats. The L5SNC-induced neuropathic pain also involves high mobility group box 1 (HMGB1), a damage-associated molecular pattern protein, and its target, the receptor for advanced glycation end-products (RAGE). We thus studied the molecular mechanisms for the L5SNC-induced Cav3.2 overexpression as well as neuropathic pain in rats by focusing on; 1) possible involvement of early growth response 1 (Egr-1), known to regulate transcriptional expression of Cav3.2, and ubiquitin-specific protease 5 (USP5) that protects Cav3.2 from proteasomal degradation, and 2) possible role of HMGB1/RAGE as an upstream signal. Protein levels of Cav3.2 as well as Egr-1 in L4 DRG significantly increased in the early (day 6) and persistent (day 14) phases of neuropathy after L5SNC, while USP5 protein in L4 DRG did not increase on day 6, but day 14. An anti-HMGB1-neutralizing antibody or a low molecular weight heparin, a RAGE antagonist, prevented the development of neuropathic pain and upregulation of Egr-1 and Cav3.2 in L4 DRG after L5SNC. L5SNC increased macrophages accumulating in the sciatic nerves, and the cytoplasm/nuclear ratio of immunoreactive HMGB1 in those macrophages. Our findings suggest that L5SNC-induced Cav3.2 overexpression in L4 DRG and neuropathic pain involves Egr-1 upregulation downstream of the macrophage-derived HMGB1/RAGE pathway, and that the delayed upregulation of USP5 might contribute to the persistent Cav3.2 overexpression and neuropathy.


Assuntos
Canais de Cálcio Tipo T/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Gânglios Espinais/metabolismo , Proteína HMGB1/biossíntese , Neuralgia/metabolismo , Proteases Específicas de Ubiquitina/biossíntese , Animais , Canais de Cálcio Tipo T/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Gânglios Espinais/patologia , Expressão Gênica , Proteína HMGB1/genética , Vértebras Lombares , Masculino , Neuralgia/genética , Neuralgia/patologia , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Nervos Espinhais/lesões , Nervos Espinhais/metabolismo , Nervos Espinhais/patologia , Proteases Específicas de Ubiquitina/genética
7.
Exp Neurol ; 317: 226-243, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30872136

RESUMO

The T-type calcium channels Cav3.2, one of the low voltage-activated (LVA) calcium channels, have been found to play important roles in the neuronal excitability. Recently, we and others have demonstrated that accumulation of Cav3.2 channels in the dorsal root ganglion (DRG) neurons and sensory nerves contributes to neuropathic pain after peripheral nerve injury. In the present study, we aimed to further investigate the regulation of Cav3.2 channels by interleukin-6 (IL-6) in DRG neurons in neuropathic pain rats after spinal nerve ligation (SNL). The results showed that Cav3.2 channel protein expression in L5 DRG neurons was upregulated and blockade of this channel decreased the hyperexcitability of DRG neurons and mechanical allodynia in SNL neuropathic pain rats. Furthermore, inhibition of IL-6 trans-signaling reduced the upregulation of Cav3.2 T-type channel induced by FIL-6 (a fusion protein of IL-6 and sIL-6R) in primary cultured DRG neurons in vitro. In vivo, inhibition of IL-6 trans-signaling reversed the upregulation of Cav3.2, reduced the hyperexcitability of L5 DRG neurons and alleviated mechanical allodynia in SNL rats. Our results suggest that IL-6 upregulates Cav3.2 T-type channels expression and function through the IL-6/sIL-6R trans-signaling pathway in DRG neurons, thus contributes to the development of neuropathic pain in SNL rats.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Gânglios Espinais/metabolismo , Interleucina-6/biossíntese , Neuralgia/metabolismo , Neurônios/metabolismo , Nervos Espinhais/lesões , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Células Cultivadas , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Interleucina-6/antagonistas & inibidores , Ligadura , Masculino , Neuralgia/fisiopatologia , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Regulação para Cima
8.
Neurosci Res ; 142: 30-37, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-29684385

RESUMO

Besides the injured peripheral dorsal root ganglion (DRG) neurons, the adjacent intact DRG neurons also have important roles in neuropathic pain. Ion channels including Cav3.2 T-type calcium channel in the DRG neurons are important in the development of neuropathic pain. In the present study, we aimed to examine the expression of Cav3.2 T-type calcium channels in the intact DRG neurons in neuropathic pain. A neuropathic pain model of rat with lumbar 5 (L5) spinal nerve ligation (SNL) was established, in which the L4 DRG was separated from the axotomized L5 DRG, and the molecular, morphological and electrophysiological changes of Cav3.2 T-type calcium channels in L4 DRG neurons were investigated. Western blotting showed that total and membrane protein levels of Cav3.2 in L4 DRG neurons increased, and voltage-dependent patch clamp recordings revealed an increased T-type current density with a curve shift to the left in steady-state activation in the acutely isolated L4 DRG neurons in neuropathic pain rats. Immunofluorescent staining further showed that the membrane expression of Cav3.2 increased in CGRP-, IB4-positive small neurons and NF200-positive large ones. In conclusion, the membrane expression and the function of Cav3.2 T-type calcium channels are increased in the intact L4 DRG neurons in neuropathic pain rats with peripheral nerve injury like SNL.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Gânglios Espinais/metabolismo , Neuralgia/metabolismo , Neurônios/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Animais , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Ligadura , Vértebras Lombares , Região Lombossacral , Masculino , Neuralgia/complicações , Traumatismos dos Nervos Periféricos/complicações , Ratos Sprague-Dawley , Regulação para Cima
9.
Toxicology ; 413: 33-39, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30552955

RESUMO

Bortezomib, a first-line agent for treatment of multiple myeloma, exhibits anticancer activity through proteasome inhibition. However, bortezomib-induced peripheral neuropathy (BIPN) is one of the most serious side effects. Since decreased proteasomal degradation of Cav3.2 T-type calcium channels in the primary afferents is involved in persistent pain, we investigated whether BIPN involves increased protein levels of Cav3.2 in mice. Six repeated i.p. administrations of bortezomib for 12 days developed persistent mechanical allodynia. Systemic administration of novel T-type calcium channel blockers, (2R/S)-6-prenylnaringenin and KTt-45, and of TTA-A2, the well-known blocker, reversed the BIPN. Ascorbic acid, known to block Cav3.2, but not Cav3.1 or 3.3, and silencing of Cav3.2 gene also suppressed BIPN. Protein levels of Cav3.2 in the dorsal root ganglion (DRG) at L4-L6 levels increased throughout days 1-21 after the onset of bortezomib treatment. Protein levels of USP5, a deubiquitinating enzyme that specifically inhibits proteasomal degradation of Cav3.2, increased in DRG on days 3-21, but not day 1, in bortezomib-treated mice. In DRG-derived ND7/23 cells, bortezomib increased protein levels of Cav3.2 and T-channel-dependent currents, as assessed by a patch-clamp method, but did not upregulate expression of Cav3.2 mRNA or USP5 protein. MG-132, another proteasome inhibitor, also increased Cav3.2 protein levels in the cultured cells. Given the previous evidence for USP5 induction following nociceptor excitation, our data suggest that BIPN involves the increased protein levels of Cav3.2 in nociceptors through inhibition of proteasomal degradation of Cav3.2 by bortezomib itself and then by USP5 that is upregulated probably in an activity-dependent manner.


Assuntos
Antineoplásicos/toxicidade , Bortezomib/toxicidade , Canais de Cálcio Tipo T/biossíntese , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/metabolismo , Inibidores de Proteassoma/toxicidade , Animais , Canais de Cálcio Tipo T/deficiência , Canais de Cálcio Tipo T/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Técnicas de Silenciamento de Genes/métodos , Masculino , Camundongos , Doenças do Sistema Nervoso Periférico/genética , Ratos
10.
Toxicology ; 393: 102-112, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29129814

RESUMO

Cav3.2 T-type Ca2+ channel activity is suppressed by zinc that binds to the extracellular histidine-191 of Cav3.2, and enhanced by H2S that interacts with zinc. Cav3.2 in nociceptors is upregulated in an activity-dependent manner. The enhanced Cav3.2 activity by H2S formed by the upregulated cystathionine-γ-lyase (CSE) is involved in the cyclophosphamide (CPA)-induced cystitis-related bladder pain in mice. We thus asked if zinc deficiency affects the cystitis-related bladder pain in mice by altering Cav3.2 function and/or expression. Dietary zinc deficiency for 2 weeks greatly decreased zinc concentrations in the plasma but not bladder tissue, and enhanced the bladder pain/referred hyperalgesia (BP/RH) following CPA at 200mg/kg, a subeffective dose, but not 400mg/kg, a maximal dose, an effect abolished by pharmacological blockade or gene silencing of Cav3.2. Acute zinc deficiency caused by systemic N,N,N',N'-tetrakis-(2-pyridylmethyl)-ethylendiamine (TPEN), a zinc chelator, mimicked the dietary zinc deficiency-induced Cav3.2-dependent promotion of BP/RH following CPA at 200mg/kg. CPA at 400mg/kg alone or TPEN plus CPA at 200mg/kg caused Cav3.2 overexpression accompanied by upregulation of Egr-1 and USP5, known to promote transcriptional expression and reduce proteasomal degradation of Cav3.2, respectively, in the dorsal root ganglia (DRG). The CSE inhibitor, ß-cyano-l-alanine, prevented the BP/RH and upregulation of Cav3.2, Egr-1 and USP5 in DRG following TPEN plus CPA at 200mg/kg. Together, zinc deficiency promotes bladder pain accompanying CPA-induced cystitis by enhancing function and expression of Cav3.2 in nociceptors, suggesting a novel therapeutic avenue for treatment of bladder pain, such as zinc supplementation.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Cistite/metabolismo , Dor/metabolismo , Zinco/deficiência , Animais , Quelantes/farmacologia , Ciclofosfamida , Cistationina gama-Liase/metabolismo , Cistite/induzido quimicamente , Dieta , Modelos Animais de Doenças , Etilaminas , Gânglios Espinais/metabolismo , Camundongos , Oligodesoxirribonucleotídeos/farmacologia , Piridinas , Bexiga Urinária/metabolismo , Zinco/sangue , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa