Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 817
Filtrar
1.
J Physiol ; 601(13): 2621-2634, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37114864

RESUMEN

Smooth muscle voltage-gated K+ (Kv) channels in resistance arteries control vascular tone and contribute to the coupling of blood flow with local metabolic activity. Members of the Kv1 family are expressed in vascular smooth muscle and are modulated upon physiological elevation of local metabolites, including the glycolytic end-product l-lactate and superoxide-derived hydrogen peroxide (H2 O2 ). Here, we show that l-lactate elicits vasodilatation of small-diameter mesenteric arteries in a mechanism that requires lactate dehydrogenase (LDH). Using the inside-out configuration of the patch clamp technique, we show that increases in NADH that reflect LDH-mediated conversion of l-lactate to pyruvate directly stimulate the activity of single Kv1 channels and significantly enhance the sensitivity of Kv1 activity to H2 O2 . Consistent with these findings, H2 O2 -evoked vasodilatation was significantly greater in the presence of 10 mM l-lactate relative to lactate-free conditions, yet was abolished in the presence of 10 mM pyruvate, which shifts the LDH reaction towards the generation of NAD+ . Moreover, the enhancement of H2 O2 -induced vasodilatation was abolished in arteries from double transgenic mice with selective overexpression of the intracellular Kvß1.1 subunit in smooth muscle cells. Together, our results indicate that the Kvß complex of native vascular Kv1 channels serves as a nodal effector for multiple redox signals to precisely control channel activity and vascular tone in the face of dynamic tissue-derived metabolic cues. KEY POINTS: Vasodilatation of mesenteric arteries by elevated external l-lactate requires its conversion by lactate dehydrogenase. Application of either NADH or H2 O2 potentiates single Kv channel currents in excised membrane patches from mesenteric artery smooth muscle cells. The binding of NADH enhances the stimulatory effects of H2 O2 on single Kv channel activity. The vasodilatory response to H2 O2 is differentially modified upon elevation of external l-lactate or pyruvate. The presence of l-lactate enhances the vasodilatory response to H2 O2 via the Kvß subunit complex in smooth muscle.


Asunto(s)
NAD , Canales de Potasio con Entrada de Voltaje , Ratones , Animales , NAD/metabolismo , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/metabolismo , Dilatación , Canales de Potasio con Entrada de Voltaje/fisiología , Arterias Mesentéricas , Oxidación-Reducción , Piruvatos/metabolismo , Piruvatos/farmacología , Lactato Deshidrogenasas/metabolismo
3.
Elife ; 112022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35642964

RESUMEN

The KV7.4 and KV7.5 subtypes of voltage-gated potassium channels play a role in important physiological processes such as sound amplification in the cochlea and adjusting vascular smooth muscle tone. Therefore, the mechanisms that regulate KV7.4 and KV7.5 channel function are of interest. Here, we study the effect of polyunsaturated fatty acids (PUFAs) on human KV7.4 and KV7.5 channels expressed in Xenopus oocytes. We report that PUFAs facilitate activation of hKV7.5 by shifting the V50 of the conductance versus voltage (G(V)) curve toward more negative voltages. This response depends on the head group charge, as an uncharged PUFA analogue has no effect and a positively charged PUFA analogue induces positive V50 shifts. In contrast, PUFAs inhibit activation of hKV7.4 by shifting V50 toward more positive voltages. No effect on V50 of hKV7.4 is observed by an uncharged or a positively charged PUFA analogue. Thus, the hKV7.5 channel's response to PUFAs is analogous to the one previously observed in hKV7.1-7.3 channels, whereas the hKV7.4 channel response is opposite, revealing subtype-specific responses to PUFAs. We identify a unique inner PUFA interaction site in the voltage-sensing domain of hKV7.4 underlying the PUFA response, revealing an unconventional mechanism of modulation of hKV7.4 by PUFAs.


In order to carry out their roles in the body, cells need to send and receive electrical signals. They can do this by allowing ions to move in and out through dedicated pore-like structures studded through their membrane. These channels are specific to one type of ions, and their activity ­ whether they open or close ­ is carefully controlled. In humans, defective ion channels are associated with conditions such as irregular heartbeats, epileptic seizures or hearing loss. Research has identified molecules known as polyunsaturated fatty acids as being able to control the activity of certain members of the KV7 family of potassium ion channels. The KV7.1 and KV7.2/7.3 channels are respectively present in the heart and the brain; KV7.4 is important for hearing, while KV7.5 plays a key role in regulating muscle tone in blood vessels. Polyunsaturated fatty acids can activate KV7.1 and KV7.2/7.3 but their impact on KV7.4 and KV7.5 remains unclear. Frampton et al. explored this question by studying human KV7.4 and KV7.5 channels expressed in frog egg cells. This showed that fatty acids activated KV7.5 (as for KV7.1 and KV7.2/7.3), but that they reduced the activity of KV7.4. Closely examining the structure of KV7.4 revealed that the fatty acids were binding to a different region compared to the other KV7 channels. When this site was made inaccessible, fatty acids increased the activity of KV7.4, just as for the rest of the family. These results may help to understand the role of polyunsaturated fatty acids in the body. In addition, knowing how these molecules interact with channels in the same family will be useful for optimising a drug's structure to avoid side effects. However, further research will be needed to understand the broader impact in a more complex biological organism.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Ácidos Grasos Insaturados/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología
4.
Nat Commun ; 13(1): 2051, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440632

RESUMEN

Adequate oxygen delivery to the heart during stress is essential for sustaining cardiac function. Acute increases in myocardial oxygen demand evoke coronary vasodilation and enhance perfusion via functional upregulation of smooth muscle voltage-gated K+ (Kv) channels. Because this response is controlled by Kv1 accessory subunits (i.e., Kvß), which are NAD(P)(H)-dependent aldo-keto reductases, we tested the hypothesis that oxygen demand modifies arterial [NAD(H)]i, and that resultant cytosolic pyridine nucleotide redox state influences Kv1 activity. High-resolution imaging mass spectrometry and live-cell imaging reveal cardiac workload-dependent increases in NADH:NAD+ in intramyocardial arterial myocytes. Intracellular NAD(P)(H) redox ratios reflecting elevated oxygen demand potentiate native coronary Kv1 activity in a Kvß2-dependent manner. Ablation of Kvß2 catalysis suppresses redox-dependent increases in Kv1 activity, vasodilation, and the relationship between cardiac workload and myocardial blood flow. Collectively, this work suggests that the pyridine nucleotide sensitivity and enzymatic activity of Kvß2 controls coronary vasoreactivity and myocardial blood flow during metabolic stress.


Asunto(s)
NAD , Canales de Potasio con Entrada de Voltaje , Músculo Liso , Nucleótidos , Oxidación-Reducción , Oxígeno , Canales de Potasio con Entrada de Voltaje/fisiología , Piridinas
5.
Elife ; 102021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34779404

RESUMEN

Positively charged amino acids respond to membrane potential changes to drive voltage sensor movement in voltage-gated ion channels, but determining the displacements of voltage sensor gating charges has proven difficult. We optically tracked the movement of the two most extracellular charged residues (R1 and R2) in the Shaker potassium channel voltage sensor using a fluorescent positively charged bimane derivative (qBBr) that is strongly quenched by tryptophan. By individually mutating residues to tryptophan within the putative pathway of gating charges, we observed that the charge motion during activation is a rotation and a tilted translation that differs between R1 and R2. Tryptophan-induced quenching of qBBr also indicates that a crucial residue of the hydrophobic plug is linked to the Cole-Moore shift through its interaction with R1. Finally, we show that this approach extends to additional voltage-sensing membrane proteins using the Ciona intestinalis voltage-sensitive phosphatase (CiVSP).


Asunto(s)
Activación del Canal Iónico/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Canales de Potasio/fisiología , Animales , Fenómenos Biofísicos , Compuestos Bicíclicos Heterocíclicos con Puentes , Ciona intestinalis/enzimología , Potenciales de la Membrana , Canales de Potasio de la Superfamilia Shaker , Triptófano/química , Xenopus laevis
6.
PLoS Comput Biol ; 17(9): e1009378, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34529674

RESUMEN

Activity-dependent regulation of intrinsic excitability has been shown to greatly contribute to the overall plasticity of neuronal circuits. Such neuroadaptations are commonly investigated in patch clamp experiments using current step stimulation and the resulting input-output functions are analyzed to quantify alterations in intrinsic excitability. However, it is rarely addressed, how such changes translate to the function of neurons when they operate under natural synaptic inputs. Still, it is reasonable to expect that a strong correlation and near proportional relationship exist between static firing responses and those evoked by synaptic drive. We challenge this view by performing a high-yield electrophysiological analysis of cultured mouse hippocampal neurons using both standard protocols and simulated synaptic inputs via dynamic clamp. We find that under these conditions the neurons exhibit vastly different firing responses with surprisingly weak correlation between static and dynamic firing intensities. These contrasting responses are regulated by two intrinsic K-currents mediated by Kv1 and Kir channels, respectively. Pharmacological manipulation of the K-currents produces differential regulation of the firing output of neurons. Static firing responses are greatly increased in stuttering type neurons under blocking their Kv1 channels, while the synaptic responses of the same neurons are less affected. Pharmacological blocking of Kir-channels in delayed firing type neurons, on the other hand, exhibit the opposite effects. Our subsequent computational model simulations confirm the findings in the electrophysiological experiments and also show that adaptive changes in the kinetic properties of such currents can even produce paradoxical regulation of the firing output.


Asunto(s)
Modelos Neurológicos , Neuronas/fisiología , Potenciales de Acción/fisiología , Animales , Células Cultivadas , Biología Computacional , Simulación por Computador , Sinapsis Eléctricas/fisiología , Fenómenos Electrofisiológicos , Hipocampo/citología , Hipocampo/fisiología , Cinética , Ratones , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/fisiología
7.
Mol Brain ; 14(1): 124, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34384467

RESUMEN

To understand the role of intracellular zinc ion (Zn2+) dysregulation in mediating age-related neurodegenerative changes, particularly neurotoxicity resulting from the generation of excessive neurotoxic amyloid-ß (Aß) peptides, this study aimed to investigate whether N, N, N', N'-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN), a Zn2+-specific chelator, could attenuate Aß25-35-induced neurotoxicity and the underlying electrophysiological mechanism. We used the 3-(4, 5-dimethyl-thiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay to measure the viability of hippocampal neurons and performed single-cell confocal imaging to detect the concentration of Zn2+ in these neurons. Furthermore, we used the whole-cell patch-clamp technique to detect the evoked repetitive action potential (APs), the voltage-gated sodium and potassium (K+) channels of primary hippocampal neurons. The analysis showed that TPEN attenuated Aß25-35-induced neuronal death, reversed the Aß25-35-induced increase in intracellular Zn2+ concentration and the frequency of APs, inhibited the increase in the maximum current density of voltage-activated sodium channel currents induced by Aß25-35, relieved the Aß25-35-induced decrease in the peak amplitude of transient outward K+ currents (IA) and outward-delayed rectifier K+ currents (IDR) at different membrane potentials, and suppressed the steady-state activation and inactivation curves of IA shifted toward the hyperpolarization direction caused by Aß25-35. These results suggest that Aß25-35-induced neuronal damage correlated with Zn2+ dysregulation mediated the electrophysiological changes in the voltage-gated sodium and K+ channels. Moreover, Zn2+-specific chelator-TPEN attenuated Aß25-35-induced neuronal damage by recovering the intracellular Zn2+ concentration.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Etilenodiaminas/farmacología , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Canales de Potasio con Entrada de Voltaje/fisiología , Canales de Sodio Activados por Voltaje/fisiología , Zinc/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Femenino , Hipocampo/citología , Activación del Canal Iónico/efectos de los fármacos , Masculino , Neuronas/fisiología , Técnicas de Placa-Clamp , Ratas , Análisis de la Célula Individual
8.
Int J Oncol ; 59(4)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34414448

RESUMEN

The targeting of membrane proteins that are activated in cancer stem cells (CSCs) represents one of the key recent strategies in cancer therapy. The present study analyzed ion channel expression profiles and functions in pancreatic CSCs (PCSCs). Cells strongly expressing aldehyde dehydrogenase 1 family member A1 (ALDH1A1) were isolated from the human pancreatic PK59 cell line using fluorescence­activated cell sorting, and PCSCs were identified based on tumorsphere formation. Microarray analysis was performed to investigate the gene expression profiles in PCSCs. ALDH1A1 messenger RNA levels were higher in PCSCs compared with non­PCSCs. PCSCs were resistant to 5­fluorouracil and capable of redifferentiation. The results of the microarray analysis revealed that gene expression related to ion channels, including voltage­gated potassium channels (Kv), was upregulated in PCSCs compared with non­PCSCs. 4­Aminopyridine (4­AP), a potent Kv inhibitor, exhibited greater cytotoxicity in PCSCs compared with non­PCSCs. In a xenograft model in nude mice, tumor volumes were significantly lower in mice inoculated with PK59 cells pre­treated with 4­AP compared with those in mice injected with non­treated cells. The present results identified a role of Kv in the persistence of PCSCs and suggested that the Kv inhibitor 4­AP may have potential as a therapeutic agent for pancreatic carcinoma.


Asunto(s)
Células Madre Neoplásicas/fisiología , Neoplasias Pancreáticas/patología , Canales de Potasio con Entrada de Voltaje/fisiología , 4-Aminopiridina/farmacología , Familia de Aldehído Deshidrogenasa 1/genética , Animales , Cloruros/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Pancreáticas/tratamiento farmacológico , Retinal-Deshidrogenasa/genética
9.
J Pharmacol Sci ; 147(1): 58-61, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34294373

RESUMEN

Nicotine administration enhances object recognition memory. However, target brain regions and cellular mechanisms underlying the nicotine effects remain unclear. In mice, the novel object recognition test revealed that systemic nicotine administration before training enhanced object recognition memory. Moreover, this effect was inhibited by infusion of retigabine, a selective voltage-dependent potassium 7 (Kv7) channel opener, into the medial prefrontal cortex (mPFC) before nicotine administration. Additionally, infusion of XE-991, a selective Kv7 channel blocker, into the mPFC before training enhanced object recognition memory. Therefore, Kv7 channels in the mPFC may be at least partly involved in nicotine-induced enhancement of object recognition memory.


Asunto(s)
Memoria/efectos de los fármacos , Nicotina/farmacología , Canales de Potasio con Entrada de Voltaje/metabolismo , Corteza Prefrontal/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Animales , Antracenos/farmacología , Carbamatos/farmacología , Masculino , Ratones Endogámicos C57BL , Fenilendiaminas/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Estimulación Química
10.
Sci Rep ; 11(1): 14632, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34272451

RESUMEN

The voltage-dependent potassium channel Kv1.3 plays essential roles in the immune system, participating in leukocyte activation, proliferation and apoptosis. The regulatory subunit KCNE4 acts as an ancillary peptide of Kv1.3, modulates K+ currents and controls channel abundance at the cell surface. KCNE4-dependent regulation of the oligomeric complex fine-tunes the physiological role of Kv1.3. Thus, KCNE4 is crucial for Ca2+-dependent Kv1.3-related leukocyte functions. To better understand the role of KCNE4 in the regulation of the immune system, we manipulated its expression in various leukocyte cell lines. Jurkat T lymphocytes exhibit low KCNE4 levels, whereas CY15 dendritic cells, a model of professional antigen-presenting cells, robustly express KCNE4. When the cellular KCNE4 abundance was increased in T cells, the interaction between KCNE4 and Kv1.3 affected important T cell physiological features, such as channel rearrangement in the immunological synapse, cell growth, apoptosis and activation, as indicated by decreased IL-2 production. Conversely, ablation of KCNE4 in dendritic cells augmented proliferation. Furthermore, the LPS-dependent activation of CY15 cells, which induced Kv1.3 but not KCNE4, increased the Kv1.3-KCNE4 ratio and increased the expression of free Kv1.3 without KCNE4 interaction. Our results demonstrate that KCNE4 is a pivotal regulator of the Kv1.3 channelosome, which fine-tunes immune system physiology by modulating Kv1.3-associated leukocyte functions.


Asunto(s)
Canal de Potasio Kv1.3/fisiología , Leucocitos/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Animales , Línea Celular , Membrana Celular/metabolismo , Células Dendríticas/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad , Sinapsis Inmunológicas/fisiología , Interleucina-2/metabolismo , Activación del Canal Iónico , Células Jurkat , Ratones
11.
J Neurophysiol ; 125(5): 1954-1972, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33852808

RESUMEN

Temporal coding precision of bushy cells in the ventral cochlear nucleus (VCN), critical for sound localization and communication, depends on the generation of rapid and temporally precise action potentials (APs). Voltage-gated potassium (Kv) channels are critically involved in this. The bushy cells in rat VCN express Kv1.1, 1.2, 1.3, 1.6, 3.1, 4.2, and 4.3 subunits. The Kv1.1 subunit contributes to the generation of a temporally precise single AP. However, the understanding of the functions of other Kv subunits expressed in the bushy cells is limited. Here, we investigated the functional diversity of Kv subunits concerning their contributions to temporal coding. We characterized the electrophysiological properties of the Kv channels with different subunits using whole cell patch-clamp recording and pharmacological methods. The neuronal firing pattern changed from single to multiple APs only when the Kv1.1 subunit was blocked. The Kv subunits, including the Kv1.1, 1.2, 1.6, or 3.1, were involved in enhancing temporal coding by lowering membrane excitability, shortening AP latencies, reducing jitter, and regulating AP kinetics. Meanwhile, all the Kv subunits contributed to rapid repolarization and sharpening peaks by narrowing half-width and accelerating fall rate, and the Kv1.1 subunit also affected the depolarization of AP. The Kv1.1, 1.2, and 1.6 subunits endowed bushy cells with a rapid time constant and a low input resistance of membrane for enhancing spike timing precision. The present results indicate that the Kv channels differentially affect intrinsic membrane properties to optimize the generation of rapid and reliable APs for temporal coding.NEW & NOTEWORTHY This study investigates the roles of Kv channels in effecting precision using electrophysiological and pharmacological methods in bushy cells. Different Kv channels have varying electrophysiological characteristics, which contribute to the interplay between changes in the membrane properties and regulation of neuronal excitability which then improve temporal coding. We conclude that the Kv channels are specialized to promote the precise and rapid coding of acoustic input by optimizing the generation of reliable APs.


Asunto(s)
Potenciales de Acción/fisiología , Núcleo Coclear/fisiología , Neuronas/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Femenino , Canal de Potasio Kv.1.1/antagonistas & inhibidores , Canal de Potasio Kv.1.1/fisiología , Canal de Potasio Kv.1.2/antagonistas & inhibidores , Canal de Potasio Kv.1.2/fisiología , Canal de Potasio Kv1.6/antagonistas & inhibidores , Canal de Potasio Kv1.6/fisiología , Masculino , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
12.
J Neurosci ; 41(18): 4141-4157, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33731451

RESUMEN

Zebrafish models are used increasingly to study the molecular pathogenesis of Parkinson's disease (PD), owing to the extensive array of techniques available for their experimental manipulation and analysis. The ascending dopaminergic projection from the posterior tuberculum (TPp; diencephalic populations DC2 and DC4) to the subpallium is considered the zebrafish correlate of the mammalian nigrostriatal projection, but little is known about the neurophysiology of zebrafish DC2/4 neurons. This is an important knowledge gap, because autonomous activity in mammalian substantia nigra (SNc) dopaminergic neurons contributes to their vulnerability in PD models. Using a new transgenic zebrafish line to label living dopaminergic neurons, and a novel brain slice preparation, we conducted whole-cell patch clamp recordings of DC2/4 neurons from adult zebrafish of both sexes. Zebrafish DC2/4 neurons share many physiological properties with mammalian dopaminergic neurons, including the cell-autonomous generation of action potentials. However, in contrast to mammalian dopaminergic neurons, the pacemaker driving intrinsic rhythmic activity in zebrafish DC2/4 neurons does not involve calcium conductances, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, or sodium leak currents. Instead, voltage clamp recordings and computational models show that interactions between three components - a small, predominantly potassium, leak conductance, voltage-gated sodium channels, and voltage-gated potassium channels - are sufficient for pacemaker activity in zebrafish DC2/4 neurons. These results contribute to understanding the comparative physiology of the dopaminergic system and provide a conceptual basis for interpreting data derived from zebrafish PD models. The findings further suggest new experimental opportunities to address the role of dopaminergic pacemaker activity in the pathogenesis of PD.SIGNIFICANCE STATEMENT Posterior tuberculum (TPp) DC2/4 dopaminergic neurons are considered the zebrafish correlate of mammalian substantia nigra (SNc) neurons, whose degeneration causes the motor signs of Parkinson's disease (PD). Our study shows that DC2/4 and SNc neurons share a number of electrophysiological properties, including depolarized membrane potential, high input resistance, and continual, cell-autonomous pacemaker activity, that strengthen the basis for the increasing use of zebrafish models to study the molecular pathogenesis of PD. The mechanisms driving pacemaker activity differ between DC2/4 and SNc neurons, providing: (1) experimental opportunities to dissociate the contributions of intrinsic activity and underlying pacemaker currents to pathogenesis; and (2) essential information for the design and interpretation of studies using zebrafish PD models.


Asunto(s)
Relojes Biológicos/fisiología , Neuronas Dopaminérgicas/fisiología , Pez Cebra/fisiología , Potenciales de Acción/fisiología , Animales , Animales Modificados Genéticamente , Señalización del Calcio/fisiología , Diencéfalo/fisiología , Femenino , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Masculino , Neostriado/fisiología , Vías Nerviosas/fisiología , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/fisiología , Sustancia Negra/fisiología , Canales de Sodio Activados por Voltaje/fisiología
13.
Pediatr Res ; 89(3): 483-487, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32299088

RESUMEN

BACKGROUND: Genetic predispositions in cases suffering sudden unexpected infant death have been a research focus worldwide during the past decade. Despite large efforts, there is still uncertainty concerning the molecular pathogenesis of these deaths. With genetic technology in constant development, the possibility of an alternative approach into this research field has become available, like mRNA expression studies. METHODS: In this study, we investigated mRNA gene expression in 14 cases who died suddenly and unexpectedly from infection without a history of severe illness prior to death. The control group included eight accidents, two cases of natural death, one undetermined, one case of medical malpractice, and two homicides. The study included tissue from liver, heart, and brain using Illumina whole-genome gene expression assay. RESULTS: From the array, 19 genes showed altered expression in the infectious deaths compared to controls. Tissue from the heart showed 15 genes with altered mRNA expression compared to the control group. CONCLUSIONS: Downregulation of KCNE5 in heart tissue from cases of infectious death was of particular interest. Variants of KCNE5 are associated with Brugada syndrome and sudden death and could be responsible for the fatal outcome in the group of infectious death. IMPACT: KCNE5 is downregulated in tissue from the heart in cases of infectious death in infancy. This study provides knowledge about the gene expression profile in cases of infectious death. Variants of a gene known to give increased risk of cardiac arrhythmia is downregulated in cases of infectious death in infancy. The results could give us better knowledge as to why some infants do not survive an infection. This study provides a candidate gene for future studies.


Asunto(s)
Infecciones Bacterianas/mortalidad , Muerte Súbita/etiología , ARN Mensajero/biosíntesis , Transcriptoma , Virosis/mortalidad , Infecciones Bacterianas/genética , Estudios de Casos y Controles , Causas de Muerte , Diagnóstico Diferencial , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Lactante , Hígado/metabolismo , Masculino , Miocardio/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/fisiología , Muerte Súbita del Lactante/diagnóstico , Lóbulo Temporal/metabolismo , Análisis de Matrices Tisulares , Virosis/genética
14.
Biosci Rep ; 41(1)2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33345272

RESUMEN

OBJECTIVE: Long non-coding RNA (lncRNA) KCNQ1OT1 was reported to be tightly associated with tumorigenesis and progression of multiple cancers. However, the expression and biological functions of KCNQ1OT1 in retinoblastoma (RB) are still unknown. We aim to elucidate the potential function and underlying mechanism of KCNQ1OT1 in regulating the progression of RB. METHODS: The levels of KCNQ1OT1 were assayed by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) analysis. The cell proliferation of RB cells (Y79 and WERI-Rb-1) were evaluated through Cell Counting Kit 8 (CCK-8) assay. Meanwhile, Y79 and WERI-Rb-1 cell apoptosis and cell cycle were assessed by Flow Cytometry analysis. Dual luciferase reporter assay were performed to illustrate the interaction between KCNQ1OT1, miR-124, and SP1. RESULTS: We found that KCNQ1OT1 was up-regulated and miR-124 was down-regulated in RB tissues and cells. Moreover, knockdown of KCNQ1OT1 reduced the proliferation, migration, and cell cycle, as well as promoted cell apoptosis of Y79 and WERI-Rb-1 cells. Western blot analysis consistently proved cell cycle and apoptosis related protein expression levels. More importantly, KCNQ1OT1 was a sponge of microRNA (miR)-124. MiR-124 inhibition strongly reversed the effect on cell proliferation, cycle arrest, and apoptosis by KCNQ1OT1 knockdown mediation. In addition, KCNQ1OT1 regulated expression of SP1, a direct target of miR-124 in RB. On the other hand, miR-124 inhibitor abrogated the active effect of KCNQ1OT1 silencing on silent information regulator 1 (SIRT1)/c-Jun N-terminal kinase (JNK) signaling pathway. The function of KCNQ1OT1 was verified in vivo. CONCLUSIONS: These findings implied that KCNQ1OT1 silencing inhibited RB progression and activated SIRT1/JNK signaling pathway partially by modulating the miR-124/SP1 axis.


Asunto(s)
Movimiento Celular , Proliferación Celular , MAP Quinasa Quinasa 4/metabolismo , MicroARNs/metabolismo , Retinoblastoma/patología , Transducción de Señal , Sirtuina 1/metabolismo , Factor de Transcripción Sp1/metabolismo , Animales , Femenino , Silenciador del Gen , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/fisiología , Retinoblastoma/metabolismo
15.
Cell Physiol Biochem ; 54(4): 696-706, 2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32706220

RESUMEN

BACKGROUND/AIMS: Mutations of desmosomal genes are known to cause arrhythmogenic cardiomyopathy characterized by arrhythmias and sudden cardiac death. Previously, we described a novel genetic variant H1684R in desmoplakin gene (DSP), associated with a progressive cardiac conduction disease (PCCD). In the present study, we aimed to investigate an effect of the DSP-H1684R genetic variant on the activity of ion channels. METHODS: We used cardiomyocytes derived from induced pluripotent stem cells (iPSC cardiomyocytes) from a patient with DSP-H1684R genetic variant and from two healthy donors. Immunofluorescent staining and western blot analyses were used to characterize patient-specific cardiomyocytes. By the whole-cell voltage-clamp technique we estimated the activity of voltage-gated sodium, calcium, and potassium channels that are responsible for action potential generation and its shape. Action potentials' parameters were measured using whole-cell current-clamp technique. RESULTS: In patient-specific cardiomyocytes we observed both lower amplitudes of currents through sodium Nav1.5 channels and L-type calcium channels, but higher amplitude of current through transient-outward potassium channels in comparison to donor cardiomyocytes. Current-clamp measurements revealed shortening of action-potential in DSP-H1684R-carrying iPSC cardiomyocytes. Therefore, observed alterations in the channels activity might have a great impact on the properties of action potential and development of PCCD. CONCLUSION: Our results show that desmoplakin genetic variants, besides conduction slowing caused by structural heart remodeling, could affect multiple ion channel activity aggravating arrhythmia manifestation in PCCD.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/genética , Desmoplaquinas/genética , Bloqueo Cardíaco/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Canales Iónicos/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Canales de Calcio/fisiología , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Desmoplaquinas/metabolismo , Técnica del Anticuerpo Fluorescente , Bloqueo Cardíaco/metabolismo , Humanos , Canales Iónicos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/fisiología , Canales de Sodio Activados por Voltaje/fisiología
16.
Toxicol Appl Pharmacol ; 403: 115153, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32717242

RESUMEN

We investigated the vasodilatory effects of empagliflozin (a sodium-glucose co-transporter 2 inhibitor) and the underlying mechanisms using rabbit aorta. Empagliflozin induced vasodilation in a concentration-dependent manner independently of the endothelium. Likewise, pretreatment with the nitric oxide synthase inhibitor L-NAME or the SKca inhibitor apamin together with the IKca inhibitor TRAM-34 did not impact the vasodilatory effects of empagliflozin. Pretreatment with the adenylyl cyclase inhibitor SQ22536 or a guanylyl cyclase inhibitor ODQ or a protein kinase A (PKA) inhibitor KT5720 also did not alter the vasodilatory response of empagliflozin. However, the vasodilatory effects of empagliflozin were significantly reduced by pretreatment with the protein kinase G (PKG) inhibitor KT5823. Although application of the ATP-sensitive K+ (KATP) channel inhibitor glibenclamide, large-conductance Ca2+-activated K+ (BKCa) channel inhibitor paxilline, or inwardly rectifying K+ (Kir) channel inhibitor Ba2+ did not impact the vasodilatory effects of empagliflozin, pretreatment with the voltage-dependent K+ (Kv) channel inhibitor 4-AP reduced the vasodilatory effects of empagliflozin. Pretreatment with DPO-1 (Kv1.5 channel inhibitor), guangxitoxin (Kv2.1 channel inhibitor), or linopirdine (Kv7 channel inhibitor) had little effect on empagliflozin-induced vasodilation. Application of nifedipine (L-type Ca2+ channel inhibitor) or thapsigargin (sarco-endoplasmic reticulum Ca2+-ATPase pump inhibitor) did not impact empagliflozin-induced vasodilation. Therefore, empagliflozin induces vasodilation by activating PKG and Kv channels.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Glucósidos/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Vasodilatación/efectos de los fármacos , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Compuestos de Bencidrilo/química , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Regulación de la Expresión Génica/efectos de los fármacos , Glucósidos/química , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Estructura Molecular , Conejos , Inhibidores del Cotransportador de Sodio-Glucosa 2/química
17.
Eur J Pharmacol ; 882: 173243, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32535099

RESUMEN

This study investigated the vasodilatory effects and acting mechanism of gemigliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor. Tests were conducted in aortic rings pre-contracted with phenylephrine. Gemigliptin induced dose-dependent vasodilation of the aortic smooth muscle. Several pre-treatment groups were used to investigate the mechanism of action. While pre-treatment with paxilline, a large-conductance Ca2+-activated K+ channel inhibitor, glibenclamide, an ATP-sensitive K+ channel inhibitor, and Ba2+, an inwardly rectifying K+ channel inhibitor, had no impact on the vasodilatory effect of gemigliptin, pre-treatment with 4-aminopyridine, a voltage-dependent K+ (Kv) channel inhibitor, effectively attenuated the vasodilatory action of gemigliptin. In addition, pre-treatment with sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid significantly reduced the vasodilatory effect of gemigliptin. cAMP/PKA-related or cGMP/PKG-related signaling pathway inhibitors, including adenylyl cyclase inhibitor SQ 22536, PKA inhibitor KT 5720, guanylyl cyclase inhibitor ODQ, and PKG inhibitor KT 5823 did not alter the vasodilatory effect of gemigliptin. Similarly, elimination of the endothelium and pre-treatment with a nitric oxide (NO) synthase inhibitor (L-NAME) or small- and intermediate-conductance Ca2+-activated K+ channels (apamin and TRAM-34, respectively) did not change the gemigliptin effect. These findings suggested that gemigliptin induces vasodilation through the activation of Kv channels and SERCA pumps independent of cAMP/PKA-related or cGMP/PKG-related signaling pathways and the endothelium. Therefore, caution is required when prescribing gemigliptin to the patients with hypotension and diabetes.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Piperidonas/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Pirimidinas/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Vasodilatadores/farmacología , Animales , Aorta Torácica/fisiología , Masculino , Músculo Liso Vascular/fisiología , Conejos
18.
Inflamm Res ; 69(6): 589-598, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32306120

RESUMEN

OBJECTIVE: To evaluate the role of K+ channels in pain following gouty arthritis. METHODS: The model of acute gouty arthritis was induced by monosodium urate (MSU) in mice. The swelling degree was determined by measuring the circumference of the ankle joint. Mechanical hyperalgesia was detected by von Frey filaments. Two types of K+ currents, A-type currents (IA) and delayed rectifier currents (IK), were recorded in dorsal root ganglion (DRG) neurons using patch-clamp techniques. RESULTS: The swelling degree reached its maximum at 10 h and the minimum pain threshold was maintained between 8 and 48 h after MSU treatment in mice. The amplitudes of IA and IK in DRG neurons were moderately increased on day 1 after MSU treatment, and then, they were gradually decreased with times and reached their minimums on day 4 (for IA) or 5 (for IK). Compared with control group, the activation curve of IA was significantly shifted to more positive potential and the recovery time of IA from inactivation was markedly prolonged, but inactivation and frequency dependence of IA appeared unaffected in MSU-treated group. Additionally, no change was observed in the activation curve of IK after MSU treatment. The excitability was significantly higher in the MSU group than in the control group. CONCLUSIONS: MSU-induced gout pain may be related to the hyperexcitability of DRG neurons elicited by decreasing K+ currents.


Asunto(s)
Artritis Gotosa/fisiopatología , Dolor/fisiopatología , Canales de Potasio con Entrada de Voltaje/fisiología , Animales , Artritis Gotosa/inducido químicamente , Ganglios Espinales/fisiología , Masculino , Ratones Endogámicos ICR , Neuronas/fisiología , Dolor/inducido químicamente , Ácido Úrico
19.
J Gen Physiol ; 152(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32311044

RESUMEN

The voltage-gated potassium channel Kv1.2 plays a pivotal role in neuronal excitability and is regulated by a variety of known and unknown extrinsic factors. The canonical accessory subunit of Kv1.2, Kvß, promotes N-type inactivation and cell surface expression of the channel. We recently reported that a neutral amino acid transporter, Slc7a5, alters the function and expression of Kv1.2. In the current study, we investigated the effects of Slc7a5 on Kv1.2 in the presence of Kvß1.2 subunits. We observed that Slc7a5-induced suppression of Kv1.2 current and protein expression was attenuated with cotransfection of Kvß1.2. However, gating effects mediated by Slc7a5, including disinhibition and a hyperpolarizing shift in channel activation, were observed together with Kvß-mediated inactivation, indicating convergent regulation of Kv1.2 by both regulatory proteins. Slc7a5 influenced several properties of Kvß-induced inactivation of Kv1.2, including accelerated inactivation, a hyperpolarizing shift and greater extent of steady-state inactivation, and delayed recovery from inactivation. These modified inactivation properties were also apparent in altered deactivation of the Kv1.2/Kvß/Slc7a5 channel complex. Taken together, these findings illustrate a functional interaction arising from simultaneous regulation of Kv1.2 by Kvß and Slc7a5, leading to powerful effects on Kv1.2 expression, gating, and overall channel function.


Asunto(s)
Activación del Canal Iónico , Transportador de Aminoácidos Neutros Grandes 1 , Canales de Potasio con Entrada de Voltaje , Transportador de Aminoácidos Neutros Grandes 1/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología
20.
Neuroreport ; 31(8): 583-589, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32301815

RESUMEN

Ketamine is an intravenous anesthetic commonly used in clinical, which has sedative and analgesic effects. Potassium channels exert many physiological functions in excitable cells. Therefore, potassium channels may be one of the targets of ketamine. In this study, we used patch clamp to study the effects of ketamine on voltage-gated potassium channels in primary sensory cortex (S1) neurons. We recorded the outward potassium currents (IA) and delayed rectifier potassium currents (IK) separately. We found that ketamine both concentration-dependently inhibited IA currents and IK currents in S1 neurons. Ketamine (100 and 300 µM) induced a concentration-dependent hyperpolarizing shift in V1/2, without affecting the slope factor (κ) or inactivation of IA. Ketamine induced a concentration-dependent hyperpolarizing shift in V1/2 of IK, without affecting its κ. Ketamine (100 and 300 µM) did not alter the steady-state activation or its κ. Hence, ketamine inhibits IA and IK in a concentration-dependent manner in S1 pyramidal neurons. The inactivation of IA does not appear to be involved in the inhibitory effect of ketamine on IA. Ketamine inhibits IK mainly by speeding up the inactivation of IK in S1 pyramidal neurons.


Asunto(s)
Anestésicos Disociativos/administración & dosificación , Ketamina/administración & dosificación , Canales de Potasio con Entrada de Voltaje/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/fisiología , Animales , Potenciales de la Membrana/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA