Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Eye Res ; 206: 108543, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33744257

RESUMO

Many long non-coding RNAs (lncRNAs) can exert crucial roles in the pathogenesis of cataract, including lncRNA KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1). We aimed to further elucidate the biological role and regulatory molecular mechanism of KCNQ1OT1 in cataract. The expression of KCNQ1OT1 and miR-223-3p and BCL2 like 2 (BCL2L2) was examined by qRT-PCR. Cataract cell model was constructed by treatment with hydrogen peroxide (H2O2) in lens epithelial cells (SRA01/04). SRA01/04 cell viability and cell apoptosis were tested using CCK-8 assay and flow cytometry, respectively. Western blot (WB) was performed to measure the levels of apoptosis-related proteins and BCL2L2 protein. The oxidative stress factors were analyzed by corresponding kits. The interaction between miR-223-3p and KCNQ1OT1 or BCL2L2 was validated by dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. We found that KCNQ1OT1 was upregulated in cataract anterior lens capsule samples and H2O2-induced SRA01/04 cells. Knockdown of KCNQ1OT1 suppressed H2O2-induced SRA01/04 cell apoptosis and oxidative stress. KCNQ1OT1 acted as a sponge of miR-223-3p. Inhibition of miR-223-3p could abate the function of KCNQ1OT1 silence in H2O2-treated SRA01/04 cells. Additionally, BCL2L2 was a direct target of miR-223-3p, and miR-223-3p weakened H2O2-induced SRA01/04 cell apoptosis and oxidative stress by targeting BCL2L2. Collectively, the data suggest a role for the KCNQ1OT1/miR-223-3p/BCL2L2 axis in cataract formation but the data was generated using an epithelial cell line.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Catarata/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Estresse Oxidativo/genética , Idoso , Proteínas Reguladoras de Apoptose/biossíntese , Catarata/metabolismo , Catarata/patologia , Células Cultivadas , DNA/genética , Feminino , Humanos , Peróxido de Hidrogênio/toxicidade , Masculino , MicroRNAs/biossíntese , Pessoa de Meia-Idade , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética
2.
J Neurosci ; 37(34): 8256-8272, 2017 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-28751455

RESUMO

Dysfunction of the fast-inactivating Kv3.4 potassium current in dorsal root ganglion (DRG) neurons contributes to the hyperexcitability associated with persistent pain induced by spinal cord injury (SCI). However, the underlying mechanism is not known. In light of our previous work demonstrating modulation of the Kv3.4 channel by phosphorylation, we investigated the role of the phosphatase calcineurin (CaN) using electrophysiological, molecular, and imaging approaches in adult female Sprague Dawley rats. Pharmacological inhibition of CaN in small-diameter DRG neurons slowed repolarization of the somatic action potential (AP) and attenuated the Kv3.4 current. Attenuated Kv3.4 currents also exhibited slowed inactivation. We observed similar effects on the recombinant Kv3.4 channel heterologously expressed in Chinese hamster ovary cells, supporting our findings in DRG neurons. Elucidating the molecular basis of these effects, mutation of four previously characterized serines within the Kv3.4 N-terminal inactivation domain eliminated the effects of CaN inhibition on the Kv3.4 current. SCI similarly induced concurrent Kv3.4 current attenuation and slowing of inactivation. Although there was little change in CaN expression and localization after injury, SCI induced upregulation of the native regulator of CaN 1 (RCAN1) in the DRG at the transcript and protein levels. Consistent with CaN inhibition resulting from RCAN1 upregulation, overexpression of RCAN1 in naive DRG neurons recapitulated the effects of pharmacological CaN inhibition on the Kv3.4 current and the AP. Overall, these results demonstrate a novel regulatory pathway that links CaN, RCAN1, and Kv3.4 in DRG neurons. Dysregulation of this pathway might underlie a peripheral mechanism of pain sensitization induced by SCI.SIGNIFICANCE STATEMENT Pain sensitization associated with spinal cord injury (SCI) involves poorly understood maladaptive modulation of neuronal excitability. Although central mechanisms have received significant attention, recent studies have identified peripheral nerve hyperexcitability as a driver of persistent pain signaling after SCI. However, the ion channels and signaling molecules responsible for this change in primary sensory neuron excitability are still not well defined. To address this problem, this study used complementary electrophysiological and molecular methods to determine how Kv3.4, a voltage-gated K+ channel robustly expressed in dorsal root ganglion neurons, becomes dysfunctional upon calcineurin (CaN) inhibition. The results strongly suggest that CaN inhibition underlies SCI-induced dysfunction of Kv3.4 and the associated excitability changes through upregulation of the native regulator of CaN 1 (RCAN1).


Assuntos
Inibidores de Calcineurina/farmacologia , Calcineurina/biossíntese , Gânglios Espinais/metabolismo , Canais de Potássio Shaw/biossíntese , Traumatismos da Medula Espinal/metabolismo , Animais , Células CHO , Inibidores de Calcineurina/toxicidade , Células Cultivadas , Vértebras Cervicais , Cricetinae , Cricetulus , Feminino , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/fisiopatologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-28233058

RESUMO

In mormyrid weakly electric fish, the electric organ discharge (EOD) is used for species recognition, orientation and prey localization. Produced in the muscle-derived adult electric organ, the EOD exhibits a wide diversity across species in both waveform and duration. While certain defining EOD characteristics can be linked to anatomical features of the electric organ, many factors underlying EOD differentiation are yet unknown. Here, we report the differential expression of 13 Kv1 voltage-gated potassium channel genes, two inwardly rectifying potassium channel genes, two previously studied sodium channel genes and an ATPase pump in two sympatric species of the genus Campylomormyrus in both the adult electric organ and skeletal muscle. Campylomormyrus compressirostris displays a basal EOD, largely unchanged during development, while C. tshokwe has an elongated, putatively derived discharge. We report an upregulation in all Kv1 genes in the electric organ of Campylomormyrus tshokwe when compared to both skeletal muscle and C. compressirostris electric organ. This pattern of upregulation in a species with a derived EOD form suggests that voltage-gated potassium channels are potentially involved in the diversification of the EOD signal among mormyrid weakly electric fish.


Assuntos
Peixe Elétrico/genética , Peixe Elétrico/metabolismo , Órgão Elétrico/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Feminino , Expressão Gênica
4.
Twin Res Hum Genet ; 20(5): 389-394, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28803575

RESUMO

CDKN1C and KCNQ1OT1 are imprinted genes that might be potential regulators of placental development. This study investigated placental expressions of CDKN1C and KCNQ1OT1 in monozygotic twins with and without selective intrauterine growth restriction (sIUGR). Seventeen sIUGR and fifteen normal monozygotic(MZ) twin pairs were examined. Placental mRNA expressions of CDKN1C and KCNQ1OT1 were detected by real-time fluorescent quantitative PCR. CDKN1C protein expression was detected by immunohistochemical assay and Western-blotting. In the sIUGR group, smaller fetuses had a smaller share of the placenta, and CDKN1C protein expression was significantly increased while KCNQ1OT1 mRNA expression was significantly decreased. The CDKN1C/KCNQ1OT1 mRNA ratio was lower in the larger fetus than in the smaller fetus (p < .05). In the control group, CDKN1C protein expression showed no difference between larger and smaller fetuses, while KCNQ1OT1 mRNA expression was significantly lower in the larger fetus, and the CDKN1C/KCNQ1OT1 mRNA ratio was higher in the larger fetus than in the smaller fetus (p < .05). Our findings showed that pathogenesis of sIUGR may be related to the co-effect of the up-regulated protein expression of CDKN1C and down-regulated mRNA expression of KCNQ1OT1 in the placenta.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/biossíntese , Retardo do Crescimento Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Placenta/metabolismo , Gêmeos Monozigóticos , Adulto , Inibidor de Quinase Dependente de Ciclina p57/genética , Feminino , Retardo do Crescimento Fetal/genética , Humanos , Recém-Nascido , Masculino , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Gravidez
5.
Clin Exp Hypertens ; 37(5): 381-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25856227

RESUMO

To test the hypothesis that chronic hypoxic pulmonary hypertension (CH-PH) is associated with increased survivin and decreased voltage-gated potassium (KV) channels expression in pulmonary arteries, rats were randomized as: normoxia (N); normoxia + YM155, survivin suppressor (NY); hypoxia (H); hypoxia + YM155 (HY). HY group had significantly reduced pulmonary arterial pressure, right ventricular weight and right ventricular hypertrophy compared with H group. Survivin mRNA and protein were detected in pulmonary arteries of rats with CH-PH, but not rats without CH-PH. YM155 downregulated survivin protein and mRNA. KV channel expression and activity were upregulated after YM155 treatment. Survivin may play a role in the pathogenesis of CH-PH.


Assuntos
Regulação da Expressão Gênica , Hipertensão Pulmonar/genética , Hipóxia/genética , Imidazóis/uso terapêutico , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Músculo Liso Vascular/metabolismo , Naftoquinonas/uso terapêutico , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Doença Crônica , Modelos Animais de Doenças , Hipertensão Pulmonar/complicações , Hipertensão Pulmonar/tratamento farmacológico , Hipóxia/etiologia , Hipóxia/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Músculo Liso Vascular/fisiopatologia , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Pressão Propulsora Pulmonar , RNA/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Survivina
6.
J Biol Chem ; 287(42): 35565-35575, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22927438

RESUMO

The Kcnh1 gene encodes a voltage-gated potassium channel highly expressed in neurons and involved in tumor cell proliferation, yet its physiological roles remain unclear. We have used the zebrafish as a model to analyze Kcnh1 function in vitro and in vivo. We found that the kcnh1 gene is duplicated in teleost fish (i.e. kcnh1a and kcnh1b) and that both genes are maternally expressed during early development. In adult zebrafish, kcnh1a and kcnh1b have distinct expression patterns but share expression in brain and testis. Heterologous expression of both genes in Xenopus oocytes revealed a strong conservation of characteristic functional properties between human and fish channels, including a unique sensitivity to intracellular Ca(2+)/calmodulin and modulation of voltage-dependent gating by extracellular Mg(2+). Using a morpholino antisense approach, we demonstrate a strong kcnh1 loss-of-function phenotype in developing zebrafish, characterized by growth retardation, delayed hindbrain formation, and embryonic lethality. This late phenotype was preceded by transcriptional up-regulation of known cell-cycle inhibitors (p21, p27, cdh2) and down-regulation of pro-proliferative factors, including cyclin D1, at 70% epiboly. These results reveal an unanticipated basic activity of kcnh1 that is crucial for early embryonic development and patterning.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Transcrição Gênica/fisiologia , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/embriologia , Animais , Ciclina D1/genética , Ciclina D1/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Feminino , Humanos , Masculino , Especificidade de Órgãos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Rombencéfalo/embriologia , Xenopus laevis , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
7.
Exp Eye Res ; 116: 424-32, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24416770

RESUMO

Human, monkey, and bovine retinal pigment epithelial (RPE) cells exhibit an M-type K+ current, which in many other cell types is mediated by channels composed of KCNQ α-subunits and KCNE auxiliary subunits. Recently, we demonstrated the expression of KCNQ1, KCNQ4, and KCNQ5 in the monkey RPE. Here, we investigated the expression of KCNQ and KCNE subunits in native bovine RPE. RT-PCR analysis revealed the expression of KCNQ1, KCNQ4, and KCNQ5 transcripts in the RPE, but, in Western blot analysis of RPE plasma membranes, only KCNQ5 was detected. Among the five members of the KCNE gene family, transcripts for KCNE1, KCNE2, KCNE3, and KCNE4 were detected in bovine RPE, but only KCNE1 and KCNE2 proteins were detected. Immunohistochemistry of frozen bovine retinal sections revealed KCNE1 expression near the apical and basal membranes of the RPE, in cone outer segments, in the outer nuclear layer, and throughout the inner retina. The localization of KCNE1 in the RPE basal membrane, where KCNQ5 was previously found to be present, suggests that this ß-subunit may contribute to M-type K(+) channels in this membrane.


Assuntos
Regulação da Expressão Gênica , Canais de Potássio KCNQ/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , RNA/genética , Epitélio Pigmentado da Retina/metabolismo , Animais , Western Blotting , Bovinos , Imuno-Histoquímica , Canais de Potássio KCNQ/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Epitélio Pigmentado da Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
EMBO J ; 27(1): 168-78, 2008 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-18079696

RESUMO

Imprinting control regions (ICRs) are known to repress genes by utilizing one of two mechanisms, CTCF-mediated insulation or the transcription of non-coding RNAs (ncRNAs). The KvDMR1 ICR contains both the promoter for the Kcnq1ot1 ncRNA and two CTCF-binding sites located within sequences exhibiting repressive activity in enhancer-blocking assays. Deletion of KvDMR1 results in ubiquitous biallelic expression of eight maternal-specific genes in distal chromosome 7. Here we report that while truncation of the Kcnq1ot1 transcript results in the loss of imprinted expression of these genes in the placenta, it does not affect imprinted expression of Cdkn1c in a subset of embryonic tissues despite universal loss of paternal-specific methylation at Cdkn1c. Consistent with tissue-specific loss of imprinted expression, growth deficiency of these mutant mice was less severe than that observed previously in mice with deletion of KvDMR1. This study demonstrates that the KvDMR1 locus can silence Cdkn1c by a mechanism independent of Kcnq1ot1 transcription, perhaps by CTCF-associated repression, making it the first example of an ICR capable of silencing the same gene by two distinct mechanisms.


Assuntos
Inativação Gênica/fisiologia , Impressão Genômica/fisiologia , Proteínas de Membrana/genética , Animais , Fator de Ligação a CCCTC , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p57/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p57/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia
9.
Protein Expr Purif ; 76(2): 205-10, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21087668

RESUMO

MiRP1 (MinK related protein 1) is a membrane protein in the KCNE family. It can associate with and modulate various voltage gated potassium channels. Mutations in human MiRP1 have been found to cause many congenital and acquired long QT syndromes, which are potentially life-threatening cardiac arrhythmias. Here, human MiRP1 was over-expressed in Escherichia coli, purified and eluted into different detergents. Two dimensional (1)H-(15)N correlated solution nuclear magnetic resonance (NMR) spectra of the human MiRP1 in four different detergent micelles indicated that high resolution solution NMR spectrum can be obtained for human MiRP1 in detergent lyso-myristoylphosphatidylglycerol (LMPG). Circular dichroism (CD) spectroscopy of human MiRP1 indicated a high content of alpha-helical secondary structure in LMPG. Backbone assignments of most MiRP1 residues were achieved through a series of triple resonance NMR experiments. Secondary structure analysis based on backbone chemical shifts showed several stretches of alpha-helices along the primary sequence of MiRP1 in LMPG.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Proteínas Recombinantes/química , Dicroísmo Circular , Clonagem Molecular , Detergentes/química , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Ressonância Magnética Nuclear Biomolecular/métodos , Fosfatidilgliceróis/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/isolamento & purificação , Conformação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação
10.
Int J Biol Sci ; 17(7): 1757-1768, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33994860

RESUMO

Background: Long noncoding RNA KCNQ1 opposite strand/antisense transcript 1 (lncRNA KCNQ1OT1) is abnormally expressed in various solid tumors. The purpose of this study was to explore the prognostic value and potential functional role of lncRNA KCNQ1OT1 across cancers. Methods: We performed a meta-analysis of published literature to evaluate the prognostic value of lncRNA KCNQ1OT1 across cancers. Verification, functional analysis, and genomic variation analysis were performed using the GEPIA, TIMER, and LnCeVar databases. According to the immune cell infiltration level, we established a prognostic model of lncRNA KCNQ1OT1 expression using public datasets of TIMER. We used quantitative real-time polymerase chain reaction (RT-qPCR) and western blot to detect the expression levels of lncRNA KCNQ1OT1 and the CD155 protein in colorectal cancer (CRC) tissues and cell lines. Then, a lncRNA KCNQ1OT1-knockdown cell line was cocultured to explore the role of lncRNA KCNQ1OT1 and CD155 in the T cell response by flow cytometric analysis. Results: Our results showed that the high expression of lncRNA KCNQ1OT1 was significantly related to poor overall survival across cancers, especially CRC. Interestingly, we found that COAD patients with high lncRNA KCNQ1OT1 expression and high CD8+ T cell infiltration levels had a worse prognosis than those with low lncRNA KCNQ1OT1 expression and high CD8+ T cell infiltration levels. Moreover, lncRNA KCNQ1OT1 and CD155 showed significantly higher expression in CRC tissue than in normal tissue, and lncRNA KCNQ1OT1 expression was positively correlated with CD155 expression in CRC. Finally, knockdown of lncRNA KCNQ1OT1 reduced CD155 expression in HCT116 and SW620 cells and enhanced the immune response in coculture with CD8+ T cells. Conclusions: High lncRNA KCNQ1OT1 expression is significantly correlated with poor prognosis of CRC patients and mediates the CD8+ T cell response in CRC. These findings indicate that lncRNA KCNQ1OT1 is a prognostic biomarker and potential immune therapeutic target for enhancing the CD8+ T cell response in CRC.


Assuntos
Linfócitos T CD8-Positivos/patologia , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Apoptose , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Humanos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Células Tumorais Cultivadas
11.
Biochem Biophys Res Commun ; 395(3): 330-5, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20381460

RESUMO

AIMS: To investigate dynamic changes in the expression of HCN2, HCN4, as well as KCNE1, and KCNE2 mRNA and protein levels in ventricular cells from acute myocardial infarction (AMI) rat hearts. MAIN METHODS: An AMI model was induced by ligating the left anterior descending coronary artery (LAD) of Sprague-Dawley rats. The rats were randomly divided into four experimental groups: 24-hour (24h) post-AMI, 1-week (1w) post-AMI, 2-week (2w) post-AMI, and 4-week (4w) post-AMI; sham-operated control rat groups were established in parallel for each time point. HCN2, HCN4, KCNE1, and KCNE2 mRNA and protein levels were measured by reverse transcription-polymerase chain reaction (RT-PCR) and by immunohistochemistry and Western blot, respectively. KEY FINDINGS: Ventricular arrhythmias occurred in all the post-AMI groups, particularly in the 1w and 2w post-AMI groups. Although HCN2, HCN4, KCNE1, and KCNE2 genes were expressed in the left ventricular myocardium of sham-operated control rats, their expression increased in rat ischemic left ventricular myocardium, with dynamic changes in expression observed 4 weeks after AMI. HCN2, HCN4, and KCNE2 protein levels were highest at 1w and KCNE2 protein levels peaked at 2w post-AMI. SIGNIFICANCE: The expression of the HCN2, HCN4, as well as KCNE1, and KCNE2 genes in ventricular cells from AMI rat hearts underwent dynamic changes, reaching peak levels at 1 or 2weeks post-AMI. The increased expression maybe related to ventricular arrhythmogenesis after AMI.


Assuntos
Ventrículos do Coração/metabolismo , Canais Iônicos/biossíntese , Infarto do Miocárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio/biossíntese , Animais , Modelos Animais de Doenças , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/genética , Masculino , Canais de Potássio/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ratos , Ratos Sprague-Dawley
12.
Neuron ; 45(2): 223-32, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15664174

RESUMO

Acquisition of secondary, tertiary, and quaternary structure is critical to the fabrication, assembly, and function of ion channels, yet the relationship between these biogenic events remains unclear. We now address this issue in voltage-gated K(+) channels (Kv) for the T1 domain, an N-terminal Kv recognition domain that is responsible for subfamily-specific, efficient assembly of Kv subunits. This domain forms a 4-fold symmetric tetramer. We have identified residues along the axial T1-T1 interface that are critical for tertiary and quaternary structure, shown that mutations at one end of the axial T1 interface can perturb the crosslinking of an intersubunit cysteine pair at the other end, and demonstrated that tertiary folding and tetramerization of this Kv domain are coupled. A threshold level of tertiary folding is required for monomers to oligomerize. Coupling between tertiary and quaternary structure formation may be a common feature in the biogenesis of multimeric proteins.


Assuntos
Membrana Celular/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Dobramento de Proteína , Animais , Cisteína/química , Cães , Feminino , Modelos Moleculares , Mutação/genética , Oócitos , Estrutura Terciária de Proteína/fisiologia , Xenopus laevis
13.
Pflugers Arch ; 457(5): 1111-20, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18709386

RESUMO

This study represents an extensive characterisation of the expression and functional impact of KCNQ and KCNE accessory subunits in a murine uterus using a combination of quantitative reverse transcription polymerase chain reaction, Western blot analysis, patch clamp electrophysiology and isometric tension recording. The use of uterine tissue throughout the oestrous cycle provided a physiological model with which to assess hormonal regulation of these genes. Messenger ribonucleic acid for all KCNQ genes were detected throughout the oestrous cycle with the KCNQ1 message predominant. KCNE isoforms were detected at each stage of the cycle. KCNE4 was the most abundant (p < 0.0001), and KCNQ1, KCNQ5 and KCNE1 were up-regulated in metestrous (p < 0.0001). The K(v)7 channel inhibitor XE991 reduced outward K(+) currents and significantly increased spontaneous myometrial contractions (p < 0.05), whereas retigabine (K(v)7 activator) significantly relaxed uterine tissues (p < 0.001). These data are the first to characterise KCNQ and KCNE gene expression in a cell type outside of neurons and the cardiovascular system.


Assuntos
Ciclo Estral/fisiologia , Canal de Potássio KCNQ1/fisiologia , Miométrio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Cromanos/farmacologia , Feminino , Canal de Potássio KCNQ1/biossíntese , Canal de Potássio KCNQ1/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fenilenodiaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Sulfonamidas/farmacologia , Contração Uterina/efeitos dos fármacos
14.
Biochem Biophys Res Commun ; 384(1): 28-31, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19371729

RESUMO

Scaffolding growth factor receptor-bound (Grb) adaptor proteins are components of macromolecular signaling complexes at the plasma membrane and thus are putative regulators of ion channel activity. The present study aimed to define the impact of Grb adaptor proteins on the function of cardiac K(+) channels. To this end channel proteins were coinjected with the adaptor proteins in Xenopus oocytes and channel activity analyzed with two-electrode voltage-clamp. It is shown that coexpression of Grb adaptor proteins can reduce current amplitudes of coexpressed channels. Grb7 and 10 significantly inhibited functional currents generated by hERG, Kv1.5 and Kv4.3 channels. Only Grb10 significantly inhibited KCNQ1/KCNE1 K(+) channels, and only Grb7 reduced Kir2.3 activity, whereas neither Grb protein significantly affected the closely related Kir2.1 and Kir2.2 channels. The present observations for the first time provide evidence for a selective and modulatory role of Grb adaptor proteins in the functional expression of cardiac K(+) channels.


Assuntos
Proteína Adaptadora GRB10/metabolismo , Proteína Adaptadora GRB7/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Animais , Proteína Adaptadora GRB10/genética , Proteína Adaptadora GRB7/genética , Humanos , Miocárdio/metabolismo , Oócitos , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Transfecção , Xenopus laevis
15.
Basic Res Cardiol ; 104(6): 621-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19421833

RESUMO

Hyperpolarization-activated ion channels, encoded by four mammalian genes (HCN1-4), contribute in an important way to the cardiac pacemaker current I(f). Here, we describe the transcription profiles of the four HCN genes, the NRSF, KCNE2 and Kir2.1 genes from embryonic stage E9.5 dpc to postnatal day 120 in the mouse. Embryonic atrium and ventricle revealed abundant HCN4 transcription but other HCN transcripts were almost absent. Towards birth, HCN4 was downregulated in the atrium and almost vanished from the ventricle. After birth, however, HCN isotype transcription changed remarkably, showing increased levels of HCN1, HCN2 and HCN4 in the atrium and of HCN2 and HCN4 in the ventricle. HCN3 showed highest transcription at early embryonic stages and was hardly detectable thereafter. At postnatal day 10, HCN4 was highest in the sinoatrial node, being twofold higher than HCN1 and fivefold higher than HCN2. In the atrium, HCN4 was similar to HCN1 and sevenfold higher than HCN2. In the ventricle, in contrast, HCN2 was sixfold higher than HCN4, while HCN1 was absent. Subsequently all HCN isotype transcripts declined to lower adult levels, while ratios of HCN isotypes remained stable. In conclusion, substantial changes of HCN isotype transcription throughout cardiac development suggest that a regulated pattern of HCN isotypes is required to establish and ensure a stable heart rhythm. Furthermore, constantly low HCN transcription in adult myocardium may be required to prevent atrial and ventricular arrhythmogenesis.


Assuntos
Coração/embriologia , Coração/crescimento & desenvolvimento , Miocárdio/metabolismo , Canais de Potássio/biossíntese , Canais de Potássio/genética , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/biossíntese , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Expressão Gênica , Perfilação da Expressão Gênica , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/biossíntese , Canais Iônicos/genética , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
16.
Brain Res ; 1217: 179-84, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18511019

RESUMO

The mechanisms by which stress and electroconvulsive therapy exert opposite effects on the course of major depression are not known. Potential candidates might include the voltage-dependent potassium channels. Potassium channels play an important role in maintaining the resting membrane potential and controlling neuronal excitability. To explore this hypothesis, we examined the effects of one or several electroconvulsive stimulations and chronic restraint stress (6 h/day for 21 days) on the expression of voltage-dependent potassium channel Kv7.2, Kv11.1, and Kv11.3 mRNA in the rat brain using in situ hybridization. Repeated, but not acute, electroconvulsive stimulation increased Kv7.2 and Kv11.1 mRNA levels in the piriform cortex. In contrast, restraint stress had no significant effect on mRNA expression of Kv7.2, Kv11.1, or Kv11.3 in any of the brain regions examined. Thus, it appears that the investigated voltage-dependent potassium channels are not modulated by restraint stress at the level of mRNA expression. However, our findings suggest that repeated electroconvulsive stimulation alter Kv7.2 and Kv11.1 function in the piriform cortex, a finding with potential relevance for the chain of neurobiological events underlying the clinical effects of ECT.


Assuntos
Eletrochoque , Regulação da Expressão Gênica , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Estresse Psicológico/fisiopatologia , Animais , Expressão Gênica , Hibridização In Situ , Masculino , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Restrição Física
17.
Eur J Pharmacol ; 593(1-3): 87-91, 2008 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-18662681

RESUMO

In this study, we explored the effects of testosterone deprivation on the expression of voltage-dependent potassium (Kv) channels in vascular smooth muscle cells (VSMCs) in rats. Six months after mature male Wistar rats were castrated, functional and posttranscriptional alterations of voltage-dependent potassium channels were detected using isometric tension measurement, whole-cell patch-clamp and western blot analysis. Constriction of aortic artery rings in response to 4-aminopyridine was significantly decreased 6 months after castration. A significant decrease in the amplitude of voltage-dependent potassium currents of aortic artery smooth muscle cells was detected in castrated rats compared with control rats. The level of expression of Kv1.5 channel protein was decreased. The decreased function and suppressed Kv1.5 protein expression of Kv channels after castration were restored by testosterone replacement. We concluded that long-term deprivation of endogenous testosterone in rats significantly attenuated the function of voltage-dependent potassium channels, and that a decreased expression of Kv1.5 channel protein accounted for this alteration. Restoration of physiological concentrations of testosterone restored the impaired function of voltage-dependent potassium channels, which may provide evidence for the beneficial effects of clinical testosterone replacement.


Assuntos
Aorta Torácica/metabolismo , Orquiectomia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Testosterona/fisiologia , 4-Aminopiridina/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Separação Celular , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Testosterona/farmacologia
18.
Clin Cancer Res ; 13(3): 824-31, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17289873

RESUMO

PURPOSE: Voltage-gated Kv potassium channels, like ether a go-go (EAG) channels, have been recognized for their oncogenic potential in breast cancer and other malignant tumors. EXPERIMENTAL DESIGN: We examined the molecular and functional expression of Kv channels in human colonic cancers and colon of mice treated with the chemical carcinogens dimethylhydrazine and N-methyl-N-nitrosourea. The data were compared with results from control mice and animals with chemically induced DSS colitis. RESULTS: Electrogenic salt transport by amiloride-sensitive Na+ channels and cyclic AMP-activated cystic fibrosis transmembrane conductance regulator Cl- channels were attenuated during tumor development and colitis, whereas Ca2+-dependent transport remained unchanged. Kv channels, in particular Eag-1, were enhanced during carcinogenesis. Multiplex reverse transcription-PCR showed increased mRNA expression for Kv1.3, Kv1.5, Kv3.1, and members of the Eag channel family, after dimethylhydrazine and N-methyl-N-nitrosourea treatment. Eag-1 protein was detected in the malignant mouse colon and human colonic cancers. Genomic amplification of Eag-1 was found in 3.4% of all human colorectal adenocarcinoma and was an independent marker of adverse prognosis. CONCLUSIONS: The study predicts an oncogenic role of Kv and Eag channels for the development of colonic cancer. These channels may represent an important target for a novel pharmacotherapy of colonic cancer.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Idoso , Animais , Neoplasias da Mama , Colo/metabolismo , Diverticulite/metabolismo , Células Epiteliais/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Humanos , Hibridização in Situ Fluorescente , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química
19.
Acta Pharmacol Sin ; 29(6): 653-60, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18501111

RESUMO

AIM: Several beta-subunits have been suggested to modulate the electrophysiological properties of the transient outward current (I(to)) in cardiac myocytes, including the obligatory beta-subunit K+-channel interacting protein (KChIP2) and KCNE2. However, neither KChIP2 nor KCNE2 modulation of Kv4.x (x=2 and/or 3) can fully recapitulate the electrophysiological properties of native I(to). The present study is to investigate how I(to) current is modulated when both KChIP2 and KCNE2 are coexpressed. METHODS: Kv4.2, KChIP2c, and KCNE2 cDNA were simultaneously transfected into COS-7 cells at a molar ratio of 3:1:1. Whole-cell currents were recorded by the patch-clamp method. RESULTS: In comparison with the current regulated by KChIP2c alone, the co-expression of KCNE2 further slowed Kv4.2 current inactivation kinetics, but diminished KChIP2c-induced positive shift of the voltage-dependent activation of Kv4.2 current. Importantly, co-expression of KCNE2 accelerated the current recovery from inactivation, and caused an povershootq of peak current amplitude during Kv4.2 current recovery, a phenomenon which has been uniquely described for human I(to). However, co-expression of KCNE2 exerted no further effect on Kv4.2 current amplitude, the rate of Kv4.2 current activation and voltage-dependent inactivation. CONCLUSION: Co-expression of Kv4.2 with KChIP2c and KCNE2, but not with KChIP2c or KCNE2 alone, yields a current profile similar to native I(to). Both KChIP2c and KCNE2 simultaneously participate in recapitulation of the electrophysiological properties of I(to) in cardiac myocytes.


Assuntos
Proteínas Interatuantes com Canais de Kv/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio Shal/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Eletrofisiologia , Humanos , Cinética , Proteínas Interatuantes com Canais de Kv/biossíntese , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese
20.
Circ Res ; 96(2): 216-24, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15618540

RESUMO

Inhibition of vascular smooth muscle (VSM) delayed rectifier K+ channels (K(DR)) by 4-aminopyridine (4-AP; 200 micromol/L) or correolide (1 micromol/L), a selective inhibitor of Kv1 channels, enhanced myogenic contraction of rat mesenteric arteries (RMAs) in response to increases in intraluminal pressure. The molecular identity of K(DR) of RMA myocytes was characterized using RT-PCR, real-time PCR, and immunocytochemistry. Transcripts encoding the pore-forming Kvalpha subunits, Kv1.2, Kv1.4, Kv1.5, and Kv1.6, were identified and confirmed at the protein level with subunit-specific antibodies. Kvbeta transcript (beta1.1, beta1.2, beta1.3, and beta2.1) expression was also identified. Kv1.5 message was approximately 2-fold more abundant than that for Kv1.2 and Kv1.6. Transcripts encoding these three Kv1alpha subunits were approximately 2-fold more abundant in 1st/2nd order conduit compared with 4th order resistance RMAs, and Kvbeta1 was 8-fold higher than Kvbeta2 message. RMA K(DR) activated positive to -50 mV, exhibited incomplete inactivation, and were inhibited by 4-AP and correolide. However, neither alpha-dendrotoxin or kappa-dendrotoxin affected RMA K(DR), implicating the presence of Kv1.5 in all channels and the absence of Kv1.1, respectively. Currents mediated by channels because of coexpression of Kv1.2, Kv1.5, Kv1.6, and Kvbeta1.2 in human embryonic kidney 293 cells had biophysical and pharmacological properties similar to those of RMA K(DR). It is concluded that K(DR) channels composed of heteromultimers of Kv1 subunits play a critical role in myogenic control of arterial diameter.


Assuntos
Artérias Cerebrais/anatomia & histologia , Artérias Mesentéricas/anatomia & histologia , Músculo Liso Vascular/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Sistema Vasomotor/fisiologia , 4-Aminopiridina/farmacologia , Animais , Biopolímeros , Linhagem Celular , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Canais de Potássio de Retificação Tardia , Venenos Elapídicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Transporte de Íons/efeitos dos fármacos , Rim , Canal de Potássio Kv1.1 , Canal de Potássio Kv1.2 , Canal de Potássio Kv1.4 , Canal de Potássio Kv1.5 , Canal de Potássio Kv1.6 , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Peptídeos/farmacologia , Potássio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shab , Especificidade da Espécie , Estresse Mecânico , Triterpenos/farmacologia , Resistência Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA