Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neuropharmacology ; 185: 108399, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33400937

RESUMO

Midbrain dopamine neurons (DANs) regulate various brain functions such as motor control and motivation. Alteration of spiking activities of these neurons could contribute to severe brain disorders including Parkinson's disease and depression. Previous studies showed important roles of somatodendritic voltage-gated K+ channels (Kv) of DANs in governing neuronal excitability and dopamine release. However, it remains largely unclear about the biophysical properties and the function of Kv channels distributed at DAN axons. We performed whole-cell recordings from the axons of DANs in acute mouse midbrain and striatal slices. We detected both rapidly activating/inactivating Kv current (i.e. A-current) and rapidly activating but slowly inactivating current (i.e. D-current) in DAN axons. Pharmacological experiments with channel blockers revealed that these currents are predominantly mediated by Kv1.4 and Kv1.2 subunits, respectively. Blocking these currents could substantially prolong axonal action potentials (APs) via a reduction of their repolarization slope. Together, our results show that Kv channels mediating A- and D-currents shape AP waveforms in midbrain DAN axons, through this regulation they may control dopamine release at the axonal terminals. Therefore, these axonal Kv channels could be drug targets for brain disorders with abnormal dopamine release.


Assuntos
Potenciais de Ação/fisiologia , Axônios/fisiologia , Neurônios Dopaminérgicos/fisiologia , Canal de Potássio Kv1.3/fisiologia , Canal de Potássio Kv1.4/fisiologia , Mesencéfalo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Feminino , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/fisiologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.4/antagonistas & inibidores , Masculino , Mesencéfalo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Bloqueadores dos Canais de Potássio/farmacologia
2.
Neuropharmacology ; 177: 108248, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32736087

RESUMO

Accumulating evidence suggests a widespread role of serotonin 5-HT7 receptors (5-HT7Rs) in the physiology of cognitive and affective processing. However, we still lack insights into 5-HT7R electrophysiology. Studies analyzing the 5-HT7R-mediated changes in CA1 pyramidal neuron activity revealed that 5-HT7R activation leads to the opening of hyperpolarization-activated cyclic nucleotide-gated cation channels (HCNs). However, our group and others have shown that CA1 pyramidal cells increase their excitability following 5-HT7R activation, an effect which cannot be explained by HCN channel opening. This suggests a different ionic mechanism might be responsible. To investigate this, we performed whole-cell patch clamp recordings of CA1 pyramidal cells in rat brain slices. It was found that acute 5-HT7R activation increased membrane excitability and decreased spiking latency. Both effects were blocked by a selective 5-HT7R antagonist. Spike latency in CA1 pyramidal cells is known to be regulated by transient outward voltage-dependent A-type potassium channels. Subsequent voltage clamp recordings revealed that acute 5-HT7R activation inhibited A-type potassium currents. Pharmacological block of Kv4.2/4.3 potassium channel subunits prevented the 5-HT7R agonist-induced changes in excitability and spiking latency, whereas blocking HCN channels had no influence on these effects. Taken together, the results reveal an ionic mechanism previously not known to be associated with 5-HT7R activation. Inhibition of A-type potassium channels can fully account for increased CA1 pyramidal cell excitability after 5-HT7R activation. These results can help explain a number of behavioral and physiological findings and will hopefully lead to a better understanding of 5-HT7 receptor signaling in health and disease.


Assuntos
Região CA1 Hipocampal/metabolismo , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/metabolismo , Células Piramidais/metabolismo , Receptores de Serotonina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Masculino , Técnicas de Cultura de Órgãos , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia
3.
J Cereb Blood Flow Metab ; 40(10): 2081-2097, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31696766

RESUMO

The histone deacetylases (HDACs)-dependent mechanisms regulating gene transcription of the Na+/Ca+ exchanger isoform 3 (ncx3) after stroke are still unknown. Overexpression or knocking-down of HDAC4/HDAC5 down-regulates or increases, respectively, NCX3 mRNA and protein. Likewise, MC1568 (class IIa HDACs inhibitor), but not MS-275 (class I HDACs inhibitor) increased NCX3 promoter activity, gene and protein expression. Furthermore, HDAC4 and HDAC5 physically interacted with the transcription factor downstream regulatory element antagonist modulator (DREAM). As MC1568, DREAM knocking-down prevented HDAC4 and HDAC5 recruitment to the ncx3 promoter. Importantly, DREAM, HDAC4, and HDAC5 recruitment to the ncx3 gene was increased in the temporoparietal cortex of rats subjected to transient middle cerebral artery occlusion (tMCAO), with a consequent histone-deacetylation of ncx3 promoter. Conversely, the tMCAO-induced NCX3 reduction was prevented by intracerebroventricular injection of siDREAM, siHDAC4, and siHDAC5. Notably, MC1568 prevented oxygen glucose deprivation plus reoxygenation and tMCAO-induced neuronal damage, whereas its neuroprotective effect was abolished by ncx3 knockdown. Collectively, we found that: (1) DREAM/HDAC4/HDAC5 complex epigenetically down-regulates ncx3 gene transcription after stroke, and (2) pharmacological inhibition of class IIa HDACs reduces stroke-induced neurodetrimental effects.


Assuntos
Epigênese Genética/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Neurônios/patologia , Proteínas Repressoras/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Animais , Córtex Cerebral/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Histona Desacetilases/genética , Humanos , Hipóxia Encefálica/prevenção & controle , Infarto da Artéria Cerebral Média/patologia , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/genética , Masculino , Fármacos Neuroprotetores , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Trocador de Sódio e Cálcio/genética , Acidente Vascular Cerebral/genética
4.
Sci Rep ; 9(1): 7260, 2019 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-31086218

RESUMO

DREAM, a neuronal calcium sensor protein, has multiple cellular roles including the regulation of Ca2+ and protein homeostasis. We recently showed that reduced DREAM expression or blockade of DREAM activity by repaglinide is neuroprotective in Huntington's disease (HD). Here we used structure-based drug design to guide the identification of IQM-PC330, which was more potent and had longer lasting effects than repaglinide to inhibit DREAM in cellular and in vivo HD models. We disclosed and validated an unexplored ligand binding site, showing Tyr118 and Tyr130 as critical residues for binding and modulation of DREAM activity. IQM-PC330 binding de-repressed c-fos gene expression, silenced the DREAM effect on KV4.3 channel gating and blocked the ATF6/DREAM interaction. Our results validate DREAM as a valuable target and propose more effective molecules for HD treatment.


Assuntos
Doença de Huntington/tratamento farmacológico , Proteínas Interatuantes com Canais de Kv/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Proteínas Repressoras/efeitos dos fármacos , Animais , Sítios de Ligação , Modelos Animais de Doenças , Desenho de Fármacos , Humanos , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Camundongos , Proteínas Repressoras/antagonistas & inibidores , Relação Estrutura-Atividade
5.
Curr Neurovasc Res ; 14(4): 397-405, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29119925

RESUMO

BACKGROUND: Although the neuroprotective effect of sodium hydrosulfide (NaHS, a hydrogen sulfide donor) pretreatment has been revealed, the effect of NaHS post-conditioning remains largely unknown. OBJECTIVE: We aimed to investigate the neuroprotective effect of NaHS post-conditioning against transient Global Cerebral Ischemia (tGCI)-induced hippocampal CA1 injury and its underlying molecular mechanism. METHODS: A tGCI rat model was established using the four-vessel occlusion method for 15 min of ischemia. The survival of hippocampal neurons was determined by Nissl staining and NeuN immunostaining. Protein expression of potassium voltage-gated channel subfamily D member 2 (Kv4.2) and potassium channel interacting protein 3 (KChIP3) was assessed by Immunohistochemistry (IHC) and Western blot. RESULTS: Decreased concentrations (12 and 24 µmol/kg) of NaHS post-conditioning significantly increased the numbers of survival neurons and NeuN-positive neurons in the hippocampal CA1 region at 7 days post-tGCI (all P<0.05). NaHS post-conditioning (24 µmol/kg) at 12 and 24 hr posttGCI can achieve the best protective effect (both P<0.05). IHC data demonstrated that NaHS postconditioning (24 µmol/kg) markedly attenuated tGCI-induced down-regulation of Kv4.2 protein in the hippocampal CA1 region at 26 hr post-tGCI. Confocal images showed that Kv4.2 did not express in the neuronal nuclei but predominantly express in the neuronal dendrites. In addition, NaHS post-conditioning significantly up-regulated Kv4.2 and down-regulated KChIP3 in tGCI rats at 26 and 168 hr post- tGCI (all P<0.05). CONCLUSION: Decreased concentrations of NaHS post-conditioning at 12-24 hr post-tGCI effectively protected hippocampal CA1 neurons from tGCI-induced injury, which may be through regulating the expression of Kv4.2 and KChIP3.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Ataque Isquêmico Transitório/metabolismo , Proteínas Interatuantes com Canais de Kv/biossíntese , Neurônios/metabolismo , Canais de Potássio Shal/biossíntese , Sulfetos/uso terapêutico , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Ataque Isquêmico Transitório/tratamento farmacológico , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Wistar , Canais de Potássio Shal/agonistas , Sulfetos/farmacologia , Resultado do Tratamento
6.
Stem Cell Res ; 16(3): 568-78, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26999760

RESUMO

Little is known about the functions of downstream regulatory element antagonist modulator (DREAM) in embryonic stem cells (ESCs). However, DREAM interacts with cAMP response element-binding protein (CREB) in a Ca(2+)-dependent manner, preventing CREB binding protein (CBP) recruitment. Furthermore, CREB and CBP are involved in maintaining ESC self-renewal and pluripotency. However, a previous knockout study revealed the protective function of DREAM depletion in brain aging degeneration and that aging is accompanied by a progressive decline in stem cells (SCs) function. Interestingly, we found that DREAM is expressed in different cell types, including human ESCs (hESCs), human adipose-derived stromal cells (hASCs), human bone marrow-derived stromal cells (hBMSCs), and human newborn foreskin fibroblasts (hFFs), and that transitory inhibition of DREAM in hESCs reduces their pluripotency, increasing differentiation. We stipulate that these changes are partly mediated by increased CREB transcriptional activity. Overall, our data indicates that DREAM acts in the regulation of hESC pluripotency and could be a target to promote or prevent differentiation in embryonic cells.


Assuntos
Células-Tronco Embrionárias Humanas/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Tecido Adiposo/citologia , Antígenos de Superfície/metabolismo , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Diferenciação Celular , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação para Baixo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/genética , Proteoglicanas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Antígenos Embrionários Estágio-Específicos/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo
7.
J Gen Virol ; 92(Pt 11): 2620-2627, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21813705

RESUMO

Human papillomaviruses (HPVs) with tropism for mucosal epithelia are the major aetiological factors in cervical cancer. Most cancers are associated with so-called high-risk HPV types, in particular HPV16, and constitutive expression of the HPV16 E6 and E7 oncoproteins is critical for malignant transformation in infected keratinocytes. E6 and E7 bind to and inactivate the cellular tumour suppressors p53 and Rb, respectively, thus delaying differentiation and inducing proliferation in suprabasal keratinocytes to enable HPV replication. One member of the Rb family, p130, appears to be a particularly important target for E7 in promoting S-phase entry. Recent evidence indicates that p130 regulates cell-cycle progression as part of a large protein complex termed DREAM. The composition of DREAM is cell cycle-regulated, associating with E2F4 and p130 in G0/G1 and with the B-myb transcription factor in S/G2. In this study, we addressed whether p130-DREAM is disrupted in HPV16-transformed cervical cancer cells and whether this is a critical function for E6/E7. We found that p130-DREAM was greatly diminished in HPV16-transformed cervical carcinoma cells (CaSki and SiHa) compared with control cell lines; however, when E6/E7 expression was targeted by specific small hairpin RNAs, p130-DREAM was reformed and the cell cycle was arrested. We further demonstrated that the profound G1 arrest in E7-depleted CaSki cells was dependent on p130-DREAM reformation by also targeting the expression of the DREAM component Lin-54 and p130. The results show that continued HPV16 E6/E7 expression is necessary in cervical cancer cells to prevent cell-cycle arrest by a repressive p130-DREAM complex.


Assuntos
Ciclo Celular , Proteína Substrato Associada a Crk/metabolismo , Interações Hospedeiro-Patógeno , Papillomavirus Humano 16/patogenicidade , Proteínas Interatuantes com Canais de Kv/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteína Substrato Associada a Crk/antagonistas & inibidores , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Humanos , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Multimerização Proteica , Proteínas Repressoras/antagonistas & inibidores
8.
J Cardiovasc Pharmacol ; 56(4): 420-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20930594

RESUMO

Methadone, a synthetic opioid for treatment of chronic pain and withdrawal from opioid dependence, has been linked to QT prolongation, potentially fatal torsades de pointes, and sudden cardiac death. Concomitant use of diazepam or other benzodiazepines in methadone maintenance treatment can increase the risk of sudden death. Therefore, we determined the effects of methadone and diazepam singly and in combination on cardiac action potentials (APs) and on the major ion channels responsible for cardiac repolarization. Using patch clamp recording in human stem cell-derived cardiomyocytes and stably transfected mammalian cells, we found that methadone produced concentration-dependent AP prolongation and ion channel block at low micromolar concentrations: hERG (IC50 = 1.7 µM), hNav1.5 (11.2 µM tonic block; 5.5 µM phasic block), and hCav1.2 (26.7 µM tonic block; 7.7 µM phasic block). Methadone was less potent in hKv4.3/hKChIP2.2 (IC50 = 39.0 µM) and hKvLQT1/hminK (53.3 µM). In contrast, diazepam blocked channels only at much higher concentrations and had no effect on AP duration at 1 µM. However, coadministration of 1-µM diazepam with methadone caused a statistically significant increase in AP duration and a 4-fold attenuation of hNav1.5 block (IC50 values were 44.2 µM and 26.6 µM, respectively, for tonic and phasic block), with no significant effect on methadone-induced block of hERG, hCav1.2, hKv4.3/hKChIP2.2, and hKvLQT1/hminK channels. Thus, although diazepam alone does not prolong the QT interval, the relief of methadone-induced Na channel block may leave hERG K channel block uncompensated, thereby increasing cardiac risk.


Assuntos
Diazepam/efeitos adversos , Hipnóticos e Sedativos/efeitos adversos , Canais Iônicos/fisiologia , Metadona/efeitos adversos , Entorpecentes/efeitos adversos , Potenciais de Ação/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Células Cultivadas , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Humanos , Hipnóticos e Sedativos/farmacologia , Canais Iônicos/antagonistas & inibidores , Canal de Potássio KCNQ1/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Metadona/farmacologia , Proteínas Musculares/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Entorpecentes/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Canais de Sódio
9.
J Mol Cell Cardiol ; 45(3): 336-46, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18565539

RESUMO

The Ca(2+)-independent transient outward potassium current (I(to)) encoded by the Kv4 family of potassium channels, is central to normal repolarization of cardiac myocytes. KChIPs are a group of Ca(2+)-binding accessory subunits that modulate Kv4-encoded currents. However, the biophysical effects of KChIP2 on Kv4 currents raise questions about the role that KChIP2 plays in forming the native I(to). Previous heterologous expression studies demonstrated that the Na channel beta1 subunit modulates the gating properties of Kv4.3 to closely recapitulate native I(to) suggesting that Na(v)beta1 may modulate the function of Kv4-encoded channels in native cardiomyocytes. Therefore we hypothesized the existence of a structural or functional complex between subunits of I(to) and I(Na). In co-immunoprecipitation of proteins from neonatal rat ventricular myocardium (NRVM), Na(v)beta1 was pulled-down by Kv4.x antibodies suggesting a structural association between subunits that comprise I(to) and I(Na). Remarkably, post-transcriptional gene silencing of KChIP2 in NRVM, using small interfering RNAs specific to KChIP2, suppressed both cardiac I(to) and I(Na) consistent with a functional coupling of these channels. KChIP2 silencing suppressed Na channel alpha and beta1 subunit mRNA levels, leaving Kv4.x mRNAs unaltered, but reducing levels of immunoreactive proteins. Post-transcriptional gene silencing of Na(v)beta1 reduced its protein expression. Silencing of Na(v)beta1 also reduced mRNA and protein levels of its alpha-subunit, Na(v)1.5. Surprisingly, silencing of Na(v)beta1 also produced a reduction in KChIP2 mRNA and protein as well as Kv4.x proteins resulting in remarkably decreased I(Na) and I(to). These data are consistent with a novel structural and functional association of I(Na) and I(to) in NRVMs.


Assuntos
Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Proteínas Interatuantes com Canais de Kv/genética , Miócitos Cardíacos/metabolismo , Canais de Potássio/metabolismo , Interferência de RNA/fisiologia , Canais de Sódio/metabolismo , Sódio/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular , Células Cultivadas , Humanos , Proteínas Interatuantes com Canais de Kv/fisiologia , Técnicas de Patch-Clamp , Subunidades Proteicas/metabolismo , Ratos , Canais de Sódio/biossíntese , Canais de Sódio/genética , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem
10.
J Physiol ; 573(Pt 2): 453-68, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16581861

RESUMO

Idiopathic constipation is higher in women of reproductive age than postmenopausal women or men, suggesting that female steroid hormones influence gastrointestinal motility. How female hormones affect motility is unclear. Colonic motility is regulated by ion channels in colonic myocytes. Voltage-dependent K(+) channels serve to set the excitability of colonic muscles. We investigated regulation of Kv 4.3 channel expression in response to acute or chronic changes in female hormones. Patch clamp experiments and quantitative PCR were used to compare outward currents and transcript expression in colonic myocytes from male, non-pregnant, pregnant and ovariectomized mice. Groups of ovariectomized mice received injections of oestrogen or progesterone to investigate the effects of hormone replacement. The capacitance of colonic myocytes from non-pregnant females was larger than in males. Net outward current density in male and ovariectomized mice was higher than in non-pregnant females and oestrogen-treated ovariectomized mice. Current densities in late pregnancy were lower than in female controls. Progesterone had no effect on outward currents. A-type currents were decreased in non-pregnant females compared with ovariectomized mice, and were further decreased by pregnancy or oestrogen replacement. Kv 4.3 transcripts did not differ significantly between groups; however, expression of the potassium channel interacting protein KChIP1 was elevated in ovariectomized mice compared with female controls and oestrogen-treated ovariectomized mice. Delayed rectifier currents were not affected by oestrogen. In the mouse colon, oestrogen suppresses A-type currents, which are important for regulating excitability. These observations suggest a possible link between female hormones and altered colonic motility associated with menses, pregnancy and menopause.


Assuntos
Colo/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Proteínas Interatuantes com Canais de Kv/fisiologia , Animais , Colo/efeitos dos fármacos , Feminino , Hormônios Esteroides Gonadais/farmacologia , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ovariectomia/métodos , Gravidez
11.
Anesthesiology ; 103(4): 796-804, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16192772

RESUMO

BACKGROUND: The transient outward current Ito is an important repolarizing K current in human ventricular myocardium mediated by Kv4.3 and KChIP2.2 subunits. Inhibition of Ito by amino-amide local anesthetics may be involved in severe cardiotoxic side effects. This study elucidates the molecular mechanisms of bupivacaine interaction with complexes formed by Kv4.3 and KChIP2.2 as well as the modulatory effect of KChIP2.2. For this purpose, the pharmacologic effects of bupivacaine on Kv4.3wt/KChIP2.2 channels and on the pore mutant Kv4.3V401I were investigated. METHODS: Kv4.3/KChIP2.2 cDNA was transiently expressed in Chinese hamster ovary cells. Site-directed mutagenesis and patch clamp experiments were performed to analyze the effects of bupivacaine on wild-type and mutant channels. RESULTS: Inhibition of Kv4.3wt/KChIP2.2 channels by bupivacaine was concentration-dependent and reversible. The IC50s for inhibition of the charge conducted by Kv4.3wt/KChIP2.2 channels by bupivacaine and levobupivacaine were 55 +/- 8 and 50 +/- 5 microm, respectively. The local anesthetic accelerated macroscopic current decline of Kv4.3wt/KChIP2.2 and slowed recovery from inactivation without altering steady state inactivation. KChIP2.2 altered the response of Kv4.3wt channels to bupivacaine and bupivacaine modulated KChIP2.2 effects on Kv4.3wt channels. The pore mutation V401I slowed macroscopic current decline of Kv4.3 channels and recovery from inactivation, and it diminished modulation of gating by KChIP2.2. Bupivacaine inhibition of Kv4.3V401I resembled Kv4.3wt and was not changed by coexpression of KChIP2.2. CONCLUSIONS: These results indicate that bupivacaine blocks Kv4.3/KChIP2.2 channels from the open state. They furthermore give structural evidence that amino-amide local anesthetics interfere with the effects of KChIP2.2 on Kv4.3 by an indirect mechanism.


Assuntos
Anestésicos Locais/farmacologia , Bupivacaína/farmacologia , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Canais de Potássio Shal/antagonistas & inibidores , Animais , Células CHO , Cricetinae , Relação Dose-Resposta a Droga , Mutagênese Sítio-Dirigida , Canais de Potássio Shal/genética
12.
Expert Opin Ther Targets ; 7(2): 249-63, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12667101

RESUMO

DREAM (downstream regulatory element antagonistic modulator) was identified as a novel calcium-binding protein with pleiotropic functions in vitro that are as varied as that of a transcription factor, a binding partner for presenilins, and a modulator of potassium channels. This review will discuss the findings that have implicated DREAM in its various roles. As a transcriptional repressor, DREAM may control the expression of the endogenous opioid gene prodynorphin amongst others, and itself is exquisitely regulated by second messenger molecules, protein kinases and other transcription factors. Recent genetic evidence has revealed a physiological role for DREAM in pain modulation. The interplay between DREAM and prodynorphin is discussed in light of our current understanding of this Janus-like opioid gene. The potential for the involvement of DREAM in other processes beyond pain modulation is considered at the end of this review.


Assuntos
Analgésicos/farmacologia , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Dor/tratamento farmacológico , Sequência de Aminoácidos , Analgésicos/uso terapêutico , Animais , Cães , Avaliação Pré-Clínica de Medicamentos , Encefalinas/metabolismo , Previsões , Humanos , Proteínas Interatuantes com Canais de Kv/deficiência , Proteínas Interatuantes com Canais de Kv/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Dados de Sequência Molecular , Insensibilidade Congênita à Dor/genética , Canais de Potássio/fisiologia , Presenilina-2 , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Proteínas Repressoras/fisiologia , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA