Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 601
Filtrar
1.
Life Sci ; 289: 120203, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34875252

RESUMEN

OBJECTIVE: To assess the functional role of Hyperpolarization-activated cyclic nucleotide-gated gated channel (HCN) subtypes in the aging bladder phenotype characterized by diminished bladder volume sensation (BVS) with or without the detrusor instability (DI). METHODS: Expression of HCN subtypes was examined by quantitative RT-PCR and Western blot in aged male Fisher 344 rats (n = 15) and young rats (n = 15). Nocturnal urination and awake cystometry (CMG) were assessed in presence and absence of a steady state HCN channel blockade achieved with daily oral gavage of vehicle or Ivabradine (HCN blocker) 6 mg/kg for 7 days. RESULTS: The association of BVS with the age-related downregulation (~30%) of cAMP sensitive HCN1, HCN2 subtypes, and (~50%) upregulation of cAMP insensitive HCN3 subtype is evinced by the doubling in the mean urine volume of nocturnal voids (0.82 ± 0.22 mL vs 0.41 ± 0.12 mL; n = 10; p < 0.05) predicting an age-related rise in the micturition volume threshold (p < 0.0001) in CMG, which is raised further by Ivabradine treatment (p < 0.0005). Ivabradine also doubled non-voiding contractions (NVC) and maximum voiding pressure (MVP) in young and aged rats, respectively (p < 0.0001) to abolish the age-related, innate two -fold elevation in NVC not accompanied with MVP rise in untreated aged rats (p < 0.005). CONCLUSION: The age-related HCN downregulation is mechanistically linked to the exhibition of aging bladder phenotype with the manifestation of DI following steady state blockade of HCN channels in Ivabradine treated young rats. The amplification of MVP in aged rats mediated by FDA approved Ivabradine hints at potential repurposing opportunity in detrusor underactivity.


Asunto(s)
Envejecimiento/metabolismo , Regulación de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales de Potasio/biosíntesis , Vejiga Urinaria de Baja Actividad/metabolismo , Vejiga Urinaria/metabolismo , Envejecimiento/patología , Animales , Masculino , Ratas , Ratas Endogámicas F344 , Vejiga Urinaria/patología , Vejiga Urinaria de Baja Actividad/patología
2.
Mol Neurobiol ; 58(7): 3575-3587, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33772465

RESUMEN

Peripheral neuropathy is the most frequent dose-limiting adverse effect of oxaliplatin. Acute pain symptoms that are induced or exacerbated by cold occur in almost all patients immediately following the first infusions. Evidence has shown that oxaliplatin causes ion channel expression modulations in dorsal root ganglia neurons, which are thought to contribute to peripheral hypersensitivity. Most dysregulated genes encode ion channels involved in cold and mechanical perception, noteworthy members of a sub-group of potassium channels of the K2P family, TREK and TRAAK. Downregulation of these K2P channels has been identified as an important tuner of acute oxaliplatin-induced hypersensitivity. We investigated the molecular mechanisms underlying this peripheral dysregulation in a murine model of neuropathic pain triggered by a single oxaliplatin administration. We found that oxaliplatin-mediated TREK-TRAAK downregulation, as well as downregulation of other K+ channels of the K2P and Kv families, involves a transcription factor known as the neuron-restrictive silencer factor (NRSF) and its epigenetic co-repressors histone deacetylases (HDACs). NRSF knockdown was able to prevent most of these K+ channel mRNA downregulation in mice dorsal root ganglion neurons as well as oxaliplatin-induced acute cold and mechanical hypersensitivity. Interestingly, pharmacological inhibition of class I HDAC reproduces the antinociceptive effects of NRSF knockdown and leads to an increased K+ channel expression in oxaliplatin-treated mice.


Asunto(s)
Regulación hacia Abajo/fisiología , Epigénesis Genética/fisiología , Hiperalgesia/metabolismo , Oxaliplatino/toxicidad , Canales de Potasio de Dominio Poro en Tándem/biosíntesis , Transcripción Genética/fisiología , Animales , Antineoplásicos/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transcripción Genética/efectos de los fármacos
3.
J Neurosci Res ; 99(2): 699-728, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33181864

RESUMEN

Neuronal diversity in the cochlea is largely determined by ion channels. Among voltage-gated channels, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels open with hyperpolarization and depolarize the cell until the resting membrane potential. The functions for hearing are not well elucidated and knowledge about localization is controversial. We created a detailed map of subcellular location and co-expression of all four HCN subunits across different mammalian species including CBA/J, C57Bl/6N, Ly5.1 mice, guinea pigs, cats, and human subjects. We correlated age-related hearing deterioration in CBA/J and C57Bl/6N with expression levels of HCN1, -2, and -4 in individual auditory neurons from the same cohort. Spatiotemporal expression during murine postnatal development exposed HCN2 and HCN4 involvement in a critical phase of hair cell innervation. The huge diversity of subunit composition, but lack of relevant heteromeric pairing along the perisomatic membrane and axon initial segments, highlighted an active role for auditory neurons. Neuron clusters were found to be the hot spots of HCN1, -2, and -4 immunostaining. HCN channels were also located in afferent and efferent fibers of the sensory epithelium. Age-related changes on HCN subtype expression were not uniform among mice and could not be directly correlated with audiometric data. The oldest mice groups revealed HCN channel up- or downregulation, depending on the mouse strain. The unexpected involvement of HCN channels in outer hair cell function where HCN3 overlaps prestin location emphasized the importance for auditory function. A better understanding may open up new possibilities to tune neuronal responses evoked through electrical stimulation by cochlear implants.


Asunto(s)
Envejecimiento/metabolismo , Cóclea/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Neuronas/metabolismo , Canales de Potasio/fisiología , Animales , Gatos , Cóclea/crecimiento & desarrollo , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Regulación de la Expresión Génica , Cobayas , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Neuronas/ultraestructura , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Fracciones Subcelulares/metabolismo
4.
Microb Cell Fact ; 19(1): 183, 2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-32957994

RESUMEN

Resistance towards known antimalarial drugs poses a significant problem, urging for novel drugs that target vital proteins in the malaria parasite Plasmodium falciparum. However, recombinant production of malaria proteins is notoriously difficult. To address this, we have investigated two putative K+ channels, PfKch1 and PfKch2, identified in the P. falciparum genome. We show that PfKch1 and PfKch2 and a C-terminally truncated version of PfKch1 (PfKch11-1094) could indeed be functionally expressed in vivo, since a K+-uptake deficient Saccharomyces cerevisiae strain was complemented by the P. falciparum cDNAs. PfKch11-1094-GFP and GFP-PfKch2 fusion proteins were overexpressed in yeast, purified and reconstituted in lipid bilayers to determine their electrophysiological activity. Single channel conductance amounted to 16 ± 1 pS for PfKch11-1094-GFP and 28 ± 2 pS for GFP-PfKch2. We predicted regulator of K+-conductance (RCK) domains in the C-terminals of both channels, and we accordingly measured channel activity in the presence of Ca2+.


Asunto(s)
Plasmodium falciparum/genética , Canales de Potasio/biosíntesis , Proteínas Protozoarias/biosíntesis , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/metabolismo , Canales de Potasio/genética , Dominios Proteicos , Proteínas Protozoarias/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Saccharomyces cerevisiae/genética
5.
Brain Res Bull ; 159: 61-66, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32165274

RESUMEN

Our previous findings indicate that HCN2 contributes to oxaliplatin-induced neuropathic pain, but the mechanisms underlying the development of neuropathic pain are still unclear. Here, we found that the rat HCN2 levels significantly increased after high-frequency stimulation-induced long-term potentiation (LTP). Spinal local application of ZD7288 (a cyclic-nucleotide-gated-channel-specific inhibitor) prevented LTP induction after intraperitoneal injection of oxaliplatin. In addition, oxaliplatin administration induced spinal LTP via activation of the CaMKII-CREB cascade in the rat spinal dorsal horn. Moreover, we found that administration of oxaliplatin significantly increased the amplitude of excitatory postsynaptic currents and the number of action potentials, but these effects were attenuated by pretreatment with either CaMKII inhibitor KN-93 or NR2B antagonist Ro 25-6981. An increase in the phosphorylation of spinal N-methyl-d-aspartate (NMDA) receptor subunit 1 (NR1) after oxaliplatin administration was weakened by ZD7288 pretreatment. Administration of noncompetitive NMDA receptor antagonist MK-801 blocked oxaliplatin-evoked CaMKII-CREB cascade activation and prevented HCN2-mediated spinal-LTP induction in vitro and suppressed neuropathic-pain behaviors of rats. All these data suggest that HCN2 contributes to the development of neuropathic pain by inducing spinal LTP via activation of NMDA receptor-mediated CaMKII signaling.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Potenciación a Largo Plazo/fisiología , Neuralgia/metabolismo , Oxaliplatino/toxicidad , Canales de Potasio/biosíntesis , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Neuralgia/inducido químicamente , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
6.
Neurochem Int ; 125: 91-98, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30794847

RESUMEN

The Ih is a mixed depolarizing current present in neurons which, upon activation by hyperpolarization, modulates neuronal excitability in the mesocorticolimbic (MCL) system, an area which regulates emotions such as pleasure, reward, and motivation. Its biophysical properties are determined by HCN protein expression profiles, specifically HCN subunits 1-4. Previously, we reported that cocaine-induced behavioral sensitization increases HCN2 protein expression in all MCL areas with the Ventral Tegmental Area (VTA) showing the most significant increase. Recent evidence suggests that HCN4 also has an important expression in the MCL system. Although there is a significant expression of HCN channels in the MCL system their role in addictive processes is largely unknown. Thus, in this study we aim to compare HCN2 and HCN4 expression profiles and their cellular compartmental distribution in the MCL system, before and after cocaine sensitization. Surface/intracellular (S/I) ratio analysis indicates that VTA HCN2 subunits are mostly expressed in the cell surface in contrast to other areas tested. Our findings demonstrate that after cocaine sensitization, the HCN2 S/I ratio in the VTA was decreased whereas in the Prefrontal Cortex it was increased. In addition, HCN4 total expression in the VTA was decreased after cocaine sensitization, although the S/I ratio was not altered. Together, these results demonstrate differential cocaine effects on HCN2 and HCN4 protein expression profiles and therefore suggest a diverse Ih modulation of cellular activity during cocaine addictive processes.


Asunto(s)
Corteza Cerebral/metabolismo , Cocaína/farmacología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Sistema Límbico/metabolismo , Canales de Potasio/biosíntesis , Animales , Corteza Cerebral/efectos de los fármacos , Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Sistema Límbico/efectos de los fármacos , Masculino , Canales de Potasio/genética , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
7.
Physiol Res ; 68(1): 107-117, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30433806

RESUMEN

The hyperpolarization-activated cyclic-nucleotide-gated non-selective cation (HCN) channels play a potential role in the neurological basis underlying drug addiction. However, little is known about the role of HCN channels in methamphetamine (METH) abuse. In the present study, we examined the changes in working memory functions of METH re-exposed mice through Morris water maze test, and investigated the protein expression of HCN1 channels and potential mechanisms underlying the modulation of HCN channels by Western blotting analysis. Mice were injected with METH (1 mg/kg, i.p.) once per day for 6 consecutive days. After 5 days without METH, mice were re-exposed to METH at the same concentration. We found that METH re-exposure caused an enhancement of working memory, and a decrease in the HCN1 channels protein expression in both hippocampus and prefrontal cortex. The phosphorylated extracellular regulated protein kinase 1/2 (p-ERK1/2), an important regulator of HCN channels, was also obviously reduced in hippocampus and prefrontal cortex of mice with METH re-exposure. Meanwhile, acute METH exposure did not affect the working memory function and the protein expressions of HCN1 channels and p-ERK1/2. Overall, our data firstly showed the aberrant protein expression of HCN1 channels in METH re-exposed mice with enhanced working memory, which was probably related to the down-regulation of p-ERK1/2 protein expression.


Asunto(s)
Regulación hacia Abajo/fisiología , Hipocampo/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Memoria a Corto Plazo/fisiología , Metanfetamina/toxicidad , Canales de Potasio/biosíntesis , Corteza Prefrontal/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Metanfetamina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Distribución Aleatoria
8.
J Gen Physiol ; 150(8): 1189-1201, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29941431

RESUMEN

The human ether-a-go-go-related gene (hERG) encodes a voltage-gated potassium channel that controls repolarization of cardiac action potentials. Accumulating evidence suggests that most disease-related hERG mutations reduce the function of the channel by disrupting protein biogenesis of the channel in the endoplasmic reticulum (ER). However, the molecular mechanism underlying the biogenesis of ERG K+ channels is largely unknown. By forward genetic screening, we identified an ER-located chaperone CNX-1, the worm homologue of mammalian chaperone Calnexin, as a critical regulator for the protein biogenesis of UNC-103, the ERG-type K+ channel in Caenorhabditis elegans Loss-of-function mutations of cnx-1 decreased the protein level and current density of the UNC-103 K+ channel and suppressed the behavioral defects caused by a gain-of-function mutation in unc-103 Moreover, CNX-1 facilitated tetrameric assembly of UNC-103 channel subunits in a liposome-assisted cell-free translation system. Further studies showed that CNX-1 act in parallel to DNJ-1, another ER-located chaperone known to regulate maturation of UNC-103 channels, on controlling the protein biogenesis of UNC-103. Importantly, Calnexin interacted with hERG proteins in the ER in HEK293T cells. Deletion of calnexin reduced the expression and current densities of endogenous hERG K+ channels in SH-SY5Y cells. Collectively, we reveal an evolutionarily conserved chaperone CNX-1/Calnexin controlling the biogenesis of ERG-type K+ channels.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Proteínas de Unión al Calcio/fisiología , Retículo Endoplásmico/metabolismo , Canales de Potasio Éter-A-Go-Go/biosíntesis , Proteínas del Choque Térmico HSP40/fisiología , Canales de Potasio/biosíntesis , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al Calcio/genética , Calnexina/metabolismo , Células HEK293 , Proteínas del Choque Térmico HSP40/genética , Humanos , Ratones
9.
ACS Synth Biol ; 7(4): 1004-1011, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29566487

RESUMEN

Processes involved in the functional formation of prokaryotic membrane proteins have remained elusive. Here, we developed a new in vitro membrane protein expression system to detect nascent activities of the KcsA potassium channel in lipid bilayers under an applied membrane potential. The channel was synthesized using a reconstituted Escherichia coli-based in vitro transcription/translation system (IVTT) in a water-in-oil droplet lined by a membrane. The synthesized channels spontaneously incorporated into the membrane even without the translocon machinery (unassisted pathway) and formed functional channels with the correct orientation. The single-channel current of the first appearing nascent channel was captured, followed by the subsequent appearance of multiple channels. Notably, the first appearance time shortened substantially as the membrane potential was hyperpolarized. Under a steadily applied membrane potential, this system serves as a production line of membrane proteins via the unassisted pathway, mimicking the bacterial synthetic membrane.


Asunto(s)
Proteínas Bacterianas/metabolismo , Potenciales de la Membrana/fisiología , Canales de Potasio/metabolismo , Biología Sintética/métodos , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Concentración de Iones de Hidrógeno , Membrana Dobles de Lípidos , Mutación , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Ingeniería de Proteínas/instrumentación , Ingeniería de Proteínas/métodos , Biología Sintética/instrumentación
10.
Methods Mol Biol ; 1685: 171-186, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29086309

RESUMEN

Cell-free production is a valuable and alternative method for the synthesis of membrane proteins. This system offers openness allowing the researchers to modify the reaction conditions without any boundaries. Additionally, the cell-free reactions are scalable from 20 µL up to several mL, faster and suitable for the high-throughput protein production. Here, we present two cell-free systems derived from Escherichia coli (E. coli) and Spodoptera frugiperda (Sf21) lysates. In the case of the E. coli cell-free system, nanodiscs are used for the solubilization and purification of membrane proteins. In the case of the Sf21 system, endogenous microsomes with an active translocon complex are present within the lysates which facilitate the incorporation of the bacterial potassium channel KcsA within the microsomal membranes. Following cell-free synthesis, these microsomes are directly used for the functional analysis of membrane proteins.


Asunto(s)
Sistema Libre de Células , Escherichia coli/citología , Proteínas de la Membrana/biosíntesis , Animales , Escherichia coli/metabolismo , Microsomas/metabolismo , Canales de Potasio/biosíntesis , Células Sf9
11.
Nucleic Acids Res ; 45(6): 3102-3115, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-27956497

RESUMEN

The dynamic interaction of DNA methylation and transcription factor binding in regulating spatiotemporal gene expression is essential for embryogenesis, but the underlying mechanisms remain understudied. In this study, using mouse models and integration of in vitro and in vivo genetic and epigenetic analyses, we show that the binding of REST (repressor element 1 (RE1) silencing transcription factor; also known as NRSF) to its cognate RE1 sequences is temporally regulated by non-CpG methylation. This process is dependent on DNA methyltransferase 3B (DNMT3B) and leads to suppression of adult cardiac genes in developing hearts. We demonstrate that DNMT3B preferentially mediates non-CpG methylation of REST-targeted genes in the developing heart. Downregulation of DNMT3B results in decreased non-CpG methylation of RE1 sequences, reduced REST occupancy, and consequently release of the transcription suppression during later cardiac development. Together, these findings reveal a critical gene silencing mechanism in developing mammalian hearts that is regulated by the dynamic interaction of DNMT3B-mediated non-CpG methylation and REST binding.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Corazón/embriología , Miocardio/metabolismo , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Ratones , Ratones Endogámicos C57BL , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Unión Proteica , Proteínas Represoras/fisiología , ADN Metiltransferasa 3B
12.
Alcohol ; 58: 33-45, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27432260

RESUMEN

Alcohol (ethanol) dependence is a chronic relapsing brain disorder partially influenced by genetics and characterized by an inability to regulate harmful levels of drinking. Emerging evidence has linked genes that encode KV7, KIR, and KCa2 K+ channels with variation in alcohol-related behaviors in rodents and humans. This led us to experimentally test relations between K+ channel genes and escalation of drinking in a chronic-intermittent ethanol (CIE) exposure model of dependence in BXD recombinant inbred strains of mice. Transcript levels for K+ channel genes in the prefrontal cortex (PFC) and nucleus accumbens (NAc) covary with voluntary ethanol drinking in a non-dependent cohort. Transcripts that encode KV7 channels covary negatively with drinking in non-dependent BXD strains. Using a pharmacological approach to validate the genetic findings, C57BL/6J mice were allowed intermittent access to ethanol to establish baseline consumption before they were treated with retigabine, an FDA-approved KV7 channel positive modulator. Systemic administration significantly reduced drinking, and consistent with previous evidence, retigabine was more effective at reducing voluntary consumption in high-drinking than low-drinking subjects. We evaluated the specific K+ channel genes that were most sensitive to CIE exposure and identified a gene subset in the NAc and PFC that were dysregulated in the alcohol-dependent BXD cohort. CIE-induced modulation of nine genes in the NAc and six genes in the PFC covaried well with the changes in drinking induced by ethanol dependence. Here we identified novel candidate genes in the NAc and PFC that are regulated by ethanol dependence and correlate with voluntary drinking in non-dependent and dependent BXD mice. The findings that Kcnq expression correlates with drinking and that retigabine reduces consumption suggest that KV7 channels could be pharmacogenetic targets to treat individuals with alcohol addiction.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/prevención & control , Farmacogenética/métodos , Canales de Potasio/genética , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Carbamatos/uso terapéutico , Femenino , Regulación de la Expresión Génica , Masculino , Moduladores del Transporte de Membrana/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Fenilendiaminas/uso terapéutico , Canales de Potasio/biosíntesis
13.
Protein Expr Purif ; 127: 53-60, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27393071

RESUMEN

KcsA, the bacterial K(+) channel from Streptomyces lividans, is the prototypical model system to study the functional and structural correlations of the pore domain of eukaryotic voltage-gated K(+) channels (Kv channels). It contains all the molecular elements responsible for ion conduction, activation, deactivation and inactivation gating [1]. KcsA's structural simplicity makes it highly amenable for structural studies. Therefore, it is methodological advantageous to produce large amounts of functional and properly folded KcsA in a cost-effective manner. In the present study, we show an optimized protocol for the over-expression and purification of large amounts of high-quality, fully functional and crystallizable KcsA using inexpensive detergents, which significantly lowered the cost of the purification process.


Asunto(s)
Proteínas Bacterianas , Expresión Génica , Canales de Potasio , Streptomyces lividans/genética , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/metabolismo , Canales de Potasio/biosíntesis , Canales de Potasio/química , Canales de Potasio/genética , Canales de Potasio/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Streptomyces lividans/metabolismo
14.
Biochemistry ; 55(30): 4212-9, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27384110

RESUMEN

Cell free protein synthesis (CFPS) has emerged as a promising methodology for protein expression. While polypeptide production is very reliable and efficient using CFPS, the correct cotranslational folding of membrane proteins during CFPS is still a challenge. In this contribution, we describe a two-step protocol in which the integral membrane protein is initially expressed by CFPS as a precipitate followed by an in vitro folding procedure using lipid vesicles for converting the protein precipitate to the correctly folded protein. We demonstrate the feasibility of using this approach for the K(+) channels KcsA and MVP and the amino acid transporter LeuT. We determine the crystal structure of the KcsA channel obtained by CFPS and in vitro folding to show the structural similarity to the cellular expressed KcsA channel and to establish the feasibility of using this two-step approach for membrane protein production for structural studies. Our studies show that the correct folding of these membrane proteins with complex topologies can take place in vitro without the involvement of the cellular machinery for membrane protein biogenesis. This indicates that the folding instructions for these complex membrane proteins are contained entirely within the protein sequence.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/química , Canales de Potasio/biosíntesis , Canales de Potasio/química , Proteínas Bacterianas/genética , Sistema Libre de Células , Cristalografía por Rayos X , Técnicas In Vitro , Membrana Dobles de Lípidos/química , Proteínas de la Membrana/genética , Modelos Moleculares , Canales de Potasio/genética , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
15.
J Cancer Res Ther ; 12(1): 248-53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27072246

RESUMEN

AIMS: Cisplatin, a platinum-based drug, is an important weapon against many types of cancer. It is well-known that cisplatin induces apoptosis. Potassium channel plays very important role in several signaling pathways. To investigate the possibility that potassium channels also have a role in the cellular response to cisplatin, we examined the effect of cisplatin on the activity of potassium channels on CT26 cell, the colon carcinoma cell line. MATERIALS AND METHODS: The cells were cultured in DMEM, supplemented with 10< heat-inactivated fetal bovine serum. At mid-log phase, cultures were harvested, washed twice in phosphate-buffered saline, and resuspended in culture medium before use. Cells were voltage-clamped using the whole-cell patch clamp technique. Membrane current data were collected and amplified. STATISTICAL ANALYSIS: Differences between two groups were assessed by paired t-test and one sample t-test to compare the relative values. One-way ANOVA was used for all experiment with more than two groups. RESULTS: Potassium currents were detected in CT26 cells and the currents were reduced by the application of tetraethylammonium (TEA) chloride, iberiotoxin, a big conductance calcium-activated potassium channel blocker and barium. The potassium currents were enhanced to 192< by the application of cisplatin (0.5 mM). Moreover, the increase of potassium currents by cisplatin was further inhibited by the application of TEA confirming the action of cisplatin on potassium channels. In addition, relative current induced by cisplatin in CT26 cells was bit larger than in normal IEC-6 cells. CONCLUSION: Potassium currents were detected in CT26 cells and the currents were reduced by the application of tetraethylammonium (TEA) chloride, iberiotoxin, a big conductance calcium-activated potassium channel blocker and barium. The potassium currents were enhanced to 192< by the application of cisplatin (0.5 mM). Moreover, the increase of potassium currents by cisplatin was further inhibited by the application of TEA confirming the action of cisplatin on potassium channels. In addition, relative current induced by cisplatin in CT26 cells was bit larger than in normal IEC-6 cells.


Asunto(s)
Carcinoma/tratamiento farmacológico , Cisplatino/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , Canales de Potasio/biosíntesis , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Técnicas de Placa-Clamp , Péptidos/administración & dosificación , Potasio/metabolismo , Canales de Potasio/genética , Transducción de Señal/efectos de los fármacos
16.
J Neuroendocrinol ; 28(6)2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27091544

RESUMEN

The magnocellular neurosecretory cells (MNCs) of the hypothalamus secrete the neurohormones vasopressin and oxytocin. The systemic release of these hormones depends on the rate and pattern of MNC firing and it is therefore important to identify the ion channels that contribute to the electrical behaviour of MNCs. In the present study, we report evidence for the presence of Na(+) -activated K(+) (KN a ) channels in rat MNCs. KN a channels mediate outwardly rectifying K(+) currents activated by the increases in intracellular Na(+) that occur during electrical activity. Although the molecular identity of native KN a channels is unclear, their biophysical properties are consistent with those of expressed Slick (slo 2.1) and Slack (slo 2.2) proteins. Using immunocytochemistry and Western blot experiments, we found that both Slick and Slack proteins are expressed in rat MNCs. Using whole cell voltage clamp techniques on acutely isolated rat MNCs, we found that inhibiting Na(+) influx by the addition of the Na(+) channel blocker tetrodotoxin or the replacement of Na(+) in the external solution with Li(+) caused a significant decrease in sustained outward currents. Furthermore, the evoked outward current density was significantly higher in rat MNCs using patch pipettes containing 60 mm Na(+) than it was when patch pipettes containing 0 mm Na(+) were used. Our data show that functional KN a channels are expressed in rat MNCs. These channels could contribute to the activity-dependent afterhyperpolarisations that have been identified in the MNCs and thereby play a role in the regulation of their electrical behaviour.


Asunto(s)
Canales de Potasio/fisiología , Sodio/fisiología , Núcleo Supraóptico/fisiología , Animales , Células Cultivadas , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/fisiología , Litio/farmacología , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Canales de Potasio/biosíntesis , Canales de potasio activados por Sodio , Ratas , Núcleo Supraóptico/efectos de los fármacos , Tetrodotoxina/farmacología
17.
Life Sci ; 144: 94-102, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26593401

RESUMEN

AIMS: In order to determine whether klotho is involved in the therapeutic effects of Astragaloside-IV on bradycardia, we evaluated the effect of ASG-IV on klotho and the effect of klotho on HCN4 and If. MAIN METHODS: Administrating isoproterenol (5 mg/kg) for 15 days to establish a rat bradycardia model randomized SD rats into control, model (ISO) and ASG-IV (5 mg/kg/day) groups to explore the effect of ASG-IV on klotho. Rats were sacrificed on day 15 after heart rate and heart function were measured; SAN tissues were collected to measure the expression of klotho and HCN4. In vitro, neonatal rat myocardial cells were incubated with LPS for 24 h to inhibit the expression of HCN4 and incubated with LPS+ klotho to explore the effect of klotho on HCN4 expression. We also adopted full-patch-clamp technique to explore the effect of klotho on If. KEY FINDINGS: Heart rate in model group was significantly decreased (356.6±19.7 vs. 428.9±19.9 in control group, P<0.01) and ASG-IV can increase heart rate (401.4±12.0 vs. 356.6±19.7 in model group, P<0.01). The expression of klotho was also up-regulated (P<0.05). In vitro, after incubation with LPS for 24h, HCN4 expression was significantly decreased in neonatal rat myocardial cells (0.6±0.07 vs. 1.0, P<0.01) and If was significantly declined. Exogenous klotho showed protective effect on HCN4 expression (1.58±0.16 in ASG-IV group vs. 0.6±0.07 in LPS group, P<0.05) and If. SIGNIFICANCE: Klotho is involved in the treatment mechanism of ASG-IV.


Asunto(s)
Bradicardia/tratamiento farmacológico , Bradicardia/genética , Glucuronidasa/biosíntesis , Glucuronidasa/genética , Saponinas/uso terapéutico , Triterpenos/uso terapéutico , Agonistas Adrenérgicos beta , Animales , Animales Recién Nacidos , Bradicardia/inducido químicamente , Pruebas de Función Cardíaca , Frecuencia Cardíaca/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Isoproterenol , Proteínas Klotho , Lipopolisacáridos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Ratas , Ratas Sprague-Dawley , Saponinas/farmacología , Triterpenos/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
Mol Med Rep ; 13(1): 1047-53, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26648415

RESUMEN

Opioids produce delayed pre-conditioning (PC) in vivo and in vitro. Our previous research revealed that opioid­induced delayed PC has an antiapoptotic effect on pulmonary artery endothelial cells (PAECs) suffering from anoxia/reoxygenation (A/R) injury. The present study hypothesized that activation of endothelial mitochondrial ATP­sensitive potassium (KATP) channels may result in antiapoptotic effects and against dysfunction in PAECs. Cultured porcine PAECs underwent 16 h anoxia treatment, followed by 1 h reoxygenation, which occurred 24 h following pretreatment with saline (0.9% NaCl; w/v) or morphine (1 µM). To determine the underlying mechanism, a selective mitochondrial KATP inhibitor, 5­hydroxydecanoic acid (5­HD; 100 µM), and an opioid receptor antagonist, naloxone (Nal; 10 µM), were administered 30 min prior to the A/R load. The percentage of apoptotic cells was assessed by Annexin V­fluorescein isothiocyanate staining, using a fluorescence­activated cell sorter. The mRNA expression of intercellular cell adhesion molecule­1 (ICAM­1) was measured by reverse transcription­quantitative polymerase chain reaction. The endothelin­1 (ET­1) content in the supernatant of PAECs cultures was estimated by radioimmunoassay. Compared with the control, A/R caused the apoptosis of PAECs, release of ET­1 and increased mRNA expression of ICAM­1. Morphine­induced delayed PC significantly reduced PAEC apoptosis, increased the release of ET­1 and reduced the mRNA expression of ICAM­1 by ~1.7­times, compared with A/R. The protective effect of morphine was abolished by pretreatment with 5­HD and Nal, however, the two agents themselves failed to aggravate the A/R injury. These results suggested that morphine-induced delayed PC has a protective effect during A/R injury of PAECs. This effect may be mediated by mitochondrial KATP channels and is opioid receptor-dependent.


Asunto(s)
Hipoxia/genética , Morfina/administración & dosificación , Canales de Potasio/biosíntesis , Arteria Pulmonar/metabolismo , Receptores Opioides/genética , Animales , Apoptosis/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelina-1/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/patología , Molécula 1 de Adhesión Intercelular/biosíntesis , Naloxona/administración & dosificación , Canales de Potasio/genética , Arteria Pulmonar/lesiones , Arteria Pulmonar/patología , ARN Mensajero/biosíntesis , Porcinos
19.
Pharmacol Biochem Behav ; 140: 1-7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26549214

RESUMEN

Chronic cerebral hypoperfusion (CCH) causes cognitive impairments and increases the risk of Alzheimer's disease (AD) and vascular dementia (VD) through several biologically plausible pathways, yet the underlying neurobiological mechanisms are still poorly understood. In this study, we investigated whether fluoxetine, a selective serotonin reuptake inhibitor (SSRI), could play a neuroprotective role against chronic cerebral hypoperfusion injury and to clarify underlying mechanisms of its efficacy. Rats were subjected to permanent bilateral occlusion of the common carotid arteries (two-vessel occlusion, 2VO). Two weeks later, rats were treated with 30 mg/kg fluoxetine (intragastric injection, i.g.) for 6 weeks. Cognitive function was evaluated by Morris water maze (MWM) and novel objects recognition (NOR) test. Long-term potentiation (LTP) was used to address the underlying synaptic mechanisms. Western blotting was used to quantify the protein levels. Our results showed that fluoxetine treatment significantly improved the cognitive impairments caused by 2VO, accompanied with a reversion of 2VO-induced inhibitory of LTP. Furthermore, 2VO caused an up-regulation of hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) surface expressions in the hippocampal CA1 area and fluoxetine also effectively recovered the disorder of HCN2 surface expressions, which may be a possible mechanism that fluoxetine treatment ameliorates cognitive impairments in rats with CCH.


Asunto(s)
Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Trastornos Cerebrovasculares/tratamiento farmacológico , Trastornos Cerebrovasculares/fisiopatología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/fisiopatología , Fluoxetina/uso terapéutico , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales de Potasio/biosíntesis , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Animales , Región CA1 Hipocampal/irrigación sanguínea , Estenosis Carotídea/tratamiento farmacológico , Estenosis Carotídea/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/fisiopatología , Canales de Potasio/genética , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Sinapsis/efectos de los fármacos
20.
J Biol Chem ; 290(30): 18575-83, 2015 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-26100633

RESUMEN

Although recent studies have shown the sodium-activated potassium channel SLACK (KCNT1) can contribute to neuronal excitability, there remains little information on the physiological role of the closely related SLICK (KCNT2) channel. Activation of SLICK channels may be important during pathological states such as ischemia, in which an increase in intracellular sodium and chloride can perturb membrane potential and ion homeostasis. We have identified two NFκB-binding sites within the promoter region of the human SLICK (KCNT2) and orthologous rat Slick (Kcnt2) genes, suggesting that conditions in which NFκB transcriptional activity is elevated promote expression of this channel. NFκB binding to the rat Slick promoter was confirmed in vivo by ChIP analyses, and NFκB was found differentially bound to the two sites. We verified NFκB transcriptional regulation of SLICK/Slick by mutational analyses and studying gene expression by luciferase assay in P19 cells, where NFκB is constitutively active. For the rat gene, activation of the Slick promoter was found to be additive in single NFκB mutations and synergistic in double mutations. Unexpectedly, for the human gene, NFκB exhibited cooperativity in activating the SLICK promoter. The human SLICK promoter constructs were then tested under hypoxic conditions in PC-12 cells, where NFκB is not active. Only under hypoxic conditions could luciferase activity be detected; the double NFκB mutant construct failed to exhibit activity. Transcriptional regulation of Slick by NFκB was verified in primary neurons. The Slick transcript decreased 24 h after NFκB inhibition. Our data show SLICK expression is predominantly under the control of NFκB. Because neuronal NFκB activation occurs during stressful stimuli such as hypoxia and injury, our findings suggest that SLICK is a neuroprotective gene.


Asunto(s)
FN-kappa B/metabolismo , Neuronas/metabolismo , Canales de Potasio/metabolismo , Transcripción Genética , Animales , Hipoxia de la Célula/genética , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Humanos , FN-kappa B/genética , Células PC12 , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Canales de potasio activados por Sodio , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Sodio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...