Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropsychopharmacology ; 49(3): 551-560, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37660129

RESUMO

Dopaminergic signaling in the nucleus accumbens shell (NAc) regulates neuronal activity relevant to reward-related learning, including cocaine-associated behaviors. Although astrocytes respond to dopamine and cocaine with structural changes, the impact of dopamine and cocaine on astrocyte functional plasticity has not been widely studied. Specifically, behavioral implications of voltage-gated channel activity in the canonically non-excitable astrocytes are not known. We characterized potassium channel function in NAc astrocytes following exposure to exogenous dopamine or cocaine self-administration training under short (2 h/day) and extended (6 h/day) access schedules. Electrophysiological, Ca2+ imaging, mRNA, and mass spectrometry tools were used for molecular characterization. Behavioral effects were examined after NAc-targeted microinjections of channel antagonists and astroglial toxins. Exogenous dopamine increased activity of currents mediated by voltage-gated (Kv7) channels in NAc astrocytes. This was associated with a ~5-fold increase in expression of Kcnq2 transcript level in homogenized NAc micropunches. Matrix-assisted laser desorption/ionization mass spectrometry revealed increased NAc dopamine levels in extended access, relative to short access, rats. Kv7 inhibition selectively increased frequency and amplitude of astrocyte intracellular Ca2+ transients in NAc of extended access rats. Inhibition of Kv7 channels in the NAc attenuated cocaine-seeking in extended access rats only, an effect that was occluded by microinjection of the astrocyte metabolic poison, fluorocitrate. These results suggest that voltage-gated K+ channel signaling in NAc astrocytes is behaviorally relevant, support Kv7-mediated regulation of astrocyte Ca2+ signals, and propose novel mechanisms of neuroglial interactions relevant to drug use.


Assuntos
Cocaína , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Ratos , Animais , Astrócitos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Ratos Sprague-Dawley , Dopamina/farmacologia , Núcleo Accumbens
2.
J Appl Toxicol ; 44(3): 391-399, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37786982

RESUMO

The regulation of membrane potential and the contractility of vascular smooth muscle cells (VSMCs) by voltage-dependent K+ (Kv) potassium channels are well-established. In this study, native VSMCs from rabbit coronary arteries were used to investigate the inhibitory effect of sertindole, an atypical antipsychotic agent, on Kv channels. Sertindole induced dose-dependent inhibition of Kv channels, with an IC50 of 3.13 ± 0.72 µM. Although sertindole did not cause a change in the steady-state activation curve, it did lead to a negative shift in the steady-state inactivation curve. The application of 1- or 2-Hz train pulses failed to alter the sertindole-induced inhibition of Kv channels, suggesting use-independent effects of the drug. The inhibitory response to sertindole was significantly diminished by pretreatment with a Kv1.5 inhibitor but not by Kv2.1 and Kv7 subtype inhibitors. These findings demonstrate the sertindole dose-dependent and use-independent inhibition of vascular Kv channels (mainly the Kv1.5 subtype) through a mechanism that involves altering steady-state inactivation curves. Therefore, the use of sertindole as an antipsychotic drug may have adverse effects on the cardiovascular system.


Assuntos
Antipsicóticos , Imidazóis , Indóis , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Animais , Coelhos , Vasos Coronários , Antipsicóticos/toxicidade , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Bloqueadores dos Canais de Potássio/toxicidade , Miócitos de Músculo Liso
3.
Mol Psychiatry ; 28(9): 3994-4010, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37833406

RESUMO

The pathogenesis of schizophrenia is believed to involve combined dysfunctions of many proteins including microtubule-associated protein 6 (MAP6) and Kv3.1 voltage-gated K+ (Kv) channel, but their relationship and functions in behavioral regulation are often not known. Here we report that MAP6 stabilizes Kv3.1 channels in parvalbumin-positive (PV+ ) fast-spiking GABAergic interneurons, regulating behavior. MAP6-/- and Kv3.1-/- mice display similar hyperactivity and avoidance reduction. Their proteins colocalize in PV+ interneurons and MAP6 deletion markedly reduces Kv3.1 protein level. We further show that two microtubule-binding modules of MAP6 bind the Kv3.1 tetramerization domain with high affinity, maintaining the channel level in both neuronal soma and axons. MAP6 knockdown by AAV-shRNA in the amygdala or the hippocampus reduces avoidance or causes hyperactivity and recognition memory deficit, respectively, through elevating projection neuron activity. Finally, knocking down Kv3.1 or disrupting the MAP6-Kv3.1 binding in these brain regions causes avoidance reduction and hyperactivity, consistent with the effects of MAP6 knockdown. Thus, disrupting this conserved cytoskeleton-membrane interaction in fast-spiking neurons causes different degrees of functional vulnerability in various neural circuits.


Assuntos
Neurônios , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Camundongos , Animais , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Emoções , Canais de Potássio Shaw/metabolismo
4.
J Appl Toxicol ; 43(12): 1926-1933, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37551856

RESUMO

Paliperidone, an atypical antipsychotic, is widely used to treat schizophrenia. In this study, we explored whether paliperidone inhibited the voltage-dependent K+ (Kv) channels of rabbit coronary arterial smooth muscle cells. Paliperidone reduced Kv channel activity in a concentration-dependent manner with a half-maximal inhibitory concentration (IC50 ) of 16.58 ± 3.03 µM and a Hill coefficient of 0.60 ± 0.04. It did not significantly shift the steady-state activation or inactivation curves, suggesting that the drug did not affect the gating properties of Kv channels. In the presence of paliperidone, the application of 20 repetitive depolarizing pulses at 1 and 2 Hz gradually increased the inhibition of the Kv current. Further, the recovery time constant after Kv channel inactivation was increased by paliperidone, indicating that it inhibited the Kv channel in a use (state)-dependent manner. Its inhibitory effects were reduced by pretreatment with a Kv1.5 subtype inhibitor. However, pretreatment with a Kv2.1 or Kv7 inhibitor did not reduce its inhibitory effect. We conclude that paliperidone inhibits Kv channels (mainly Kv1.5 subtype channels) in a concentration- and use (state)-dependent manner without changing channel gating.


Assuntos
Antipsicóticos , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Animais , Coelhos , Antipsicóticos/toxicidade , Palmitato de Paliperidona/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Miócitos de Músculo Liso
5.
Int J Mol Sci ; 24(15)2023 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-37569465

RESUMO

Long QT syndrome (LQTS) can lead to ventricular arrhythmia and sudden cardiac death. The most common congenital cause of LQTS is mutations in the channel subunits generating the cardiac potassium current IKs. Zebrafish (Danio rerio) have been proposed as a powerful system to model human cardiac diseases due to the similar electrical properties of the zebrafish heart and the human heart. We used high-resolution all-optical electrophysiology on ex vivo zebrafish hearts to assess the effects of IKs analogues on the cardiac action potential. We found that chromanol 293B (an IKs inhibitor) prolonged the action potential duration (APD) in the presence of E4031 (an IKr inhibitor applied to drug-induced LQT2), and to a lesser extent, in the absence of E4031. Moreover, we showed that PUFA analogues slightly shortened the APD of the zebrafish heart. However, PUFA analogues failed to reverse the APD prolongation in drug-induced LQT2. However, a more potent IKs activator, ML-277, partially reversed the APD prolongation in drug-induced LQT2 zebrafish hearts. Our results suggest that IKs plays a limited role in ventricular repolarizations in the zebrafish heart under resting conditions, although it plays a more important role when the IKr is compromised, as if the IKs in zebrafish serves as a repolarization reserve as in human hearts. This study shows that potent IKs activators can restore the action potential duration in drug-induced LQT2 in the zebrafish heart.


Assuntos
Síndrome do QT Longo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Animais , Humanos , Antiarrítmicos/farmacologia , Peixe-Zebra , Coração , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/genética , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/genética , Potenciais de Ação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia
6.
Eur J Med Chem ; 259: 115561, 2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37454520

RESUMO

Voltage-gated potassium channel KV1.3 inhibitors have been shown to be effective in preventing T-cell proliferation and activation by affecting intracellular Ca2+ homeostasis. Here, we present the structure-activity relationship, KV1.3 inhibition, and immunosuppressive effects of new thiophene-based KV1.3 inhibitors with nanomolar potency on K+ current in T-lymphocytes and KV1.3 inhibition on Ltk- cells. The new KV1.3 inhibitor trans-18 inhibited KV1.3 -mediated current in phytohemagglutinin (PHA)-activated T-lymphocytes with an IC50 value of 26.1 nM and in mammalian Ltk- cells with an IC50 value of 230 nM. The KV1.3 inhibitor trans-18 also had nanomolar potency against KV1.3 in Xenopus laevis oocytes (IC50 = 136 nM). The novel thiophene-based KV1.3 inhibitors impaired intracellular Ca2+ signaling as well as T-cell activation, proliferation, and colony formation.


Assuntos
Imunossupressores , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Tiofenos , Animais , Mamíferos/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Relação Estrutura-Atividade , Linfócitos T , Tiofenos/química , Tiofenos/farmacologia , Imunossupressores/química
7.
J Appl Physiol (1985) ; 134(5): 1075-1082, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36958348

RESUMO

Slowly adapting receptors (SARs), vagal mechanosensitive receptors located in the lung, play an important role in regulating the breathing pattern and Hering-Breuer inflation reflex (HBIR). Inhalation of high concentration of sulfur dioxide (SO2), a common environmental and occupational air pollutant, has been shown to selectively block the SAR activity in rabbits, but the mechanism underlying this inhibitory effect remained a mystery. We carried out this study to determine if inhalation of SO2 can inhibit the HBIR and change the eupneic breathing pattern, and to investigate further a possible involvement of voltage-gated K+ channels in the inhibitory effect of SO2 on these vagal reflex-mediated responses. Our results showed 1) inhalation of SO2 (600 ppm; 8 min) consistently abolished both the phasic activity of SARs and their response to lung inflation in anesthetized, artificially ventilated mice, 2) inhalation of SO2 generated a distinct inhibitory effect on the HBIR and induced slow deep breathing in anesthetized, spontaneously breathing mice, and these effects were reversible and reproducible in the same animals, 3) This inhibitory effect of SO2 was blocked by pretreatment with 4-aminopyridine (4-AP), a nonselective blocker of voltage-gated K+ channel, and unaffected by pretreatment with its vehicle. In conclusion, this study suggests that this inhibitory effect on the baseline breathing pattern and the HBIR response was primarily mediated through the SO2-induced activation of voltage-gated K+ channels located in the vagal bronchopulmonary SAR neurons.NEW & NOTEWORTHY This study demonstrated that inhaled sulfur dioxide completely and reversibly abolished the activity of vagal bronchopulmonary slowly adapting receptors, significantly inhibited the apneic response to lung inflation, and induced slow deep breathing in anesthetized mice. More importantly, our results further suggested that this inhibitory effect was mediated through an action of sulfur dioxide and its derivatives on the voltage-gated potassium channels expressed in the slowly adapting receptor sensory neurons innervating the lung.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Dióxido de Enxofre , Coelhos , Animais , Camundongos , Dióxido de Enxofre/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Respiração , Pulmão , Reflexo , Nervo Vago , Apneia , 4-Aminopiridina/farmacologia
8.
Arch Toxicol ; 97(3): 849-863, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36653537

RESUMO

Exposure to endocrine-disrupting chemicals (EDCs) during development may cause reproductive disorders in women. Although female reproductive endpoints are assessed in rodent toxicity studies, a concern is that typical endpoints are not sensitive enough to detect chemicals of concern to human health. If so, measured endpoints must be improved or new biomarkers of effects included. Herein, we have characterized the dynamic transcriptional landscape of developing rat ovaries exposed to two well-known EDCs, diethylstilbestrol (DES) and ketoconazole (KTZ), by 3' RNA sequencing. Rats were orally exposed from day 7 of gestation until birth, and from postnatal day 1 until days 6, 14 or 22. Three exposure doses for each chemical were used: 3, 6 and 12 µg/kg bw/day of DES; 3, 6, 12 mg/kg bw/day of KTZ. The transcriptome changed dynamically during perinatal development in control ovaries, with 1137 differentially expressed genes (DEGs) partitioned into 3 broad expression patterns. A cross-species deconvolution strategy based on a mouse ovary developmental cell atlas was used to map any changes to ovarian cellularity across the perinatal period to allow for characterization of actual changes to gene transcript levels. A total of 184 DEGs were observed across dose groups and developmental stages in DES-exposed ovaries, and 111 DEGs in KTZ-exposed ovaries across dose groups and developmental stages. Based on our analyses, we have identified new candidate biomarkers for female reproductive toxicity induced by EDC, including Kcne2, Calb2 and Insl3.


Assuntos
Disruptores Endócrinos , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Humanos , Gravidez , Camundongos , Feminino , Ratos , Animais , Dietilestilbestrol/toxicidade , Ovário , Disruptores Endócrinos/toxicidade , Cetoconazol , Reprodução , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia
9.
Drug Chem Toxicol ; 46(2): 271-280, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35317682

RESUMO

Pimozide is an antipsychotic drug used to treat chronic psychosis, such as Tourette's syndrome. Despite its widespread clinical use, pimozide can cause unexpected adverse effects, including arrhythmias. However, the adverse effects of pimozide on vascular K+ channels have not yet been determined. Therefore, we investigated the effects of pimozide on voltage-gated K+ (Kv) channels in rabbit coronary arterial smooth muscle cells. Pimozide concentration-dependently inhibited the Kv currents with an IC50 value of 1.78 ± 0.17 µM and a Hill coefficient of 0.90 ± 0.05. The inhibitory effect on the Kv current by pimozide was highly voltage-dependent in the voltage range of Kv channel activation, and additive inhibition of the Kv current by pimozide was observed in the full activation voltage range. The decay rate of inactivation was significantly accelerated by pimozide. Pimozide shifted the inactivation curve to a more negative potential. The recovery time constant from inactivation increased in the presence of pimozide. Furthermore, pimozide-induced inhibition of the Kv current was augmented by applying train pulses. Although pretreatment with the Kv2.1 subtype inhibitor guangxitoxin and the Kv7 subtype inhibitor linopirdine did not alter the degree of pimozide-induced inhibition of the Kv currents, pretreatment with the Kv1.5 channel inhibitor DPO-1 reduced the inhibitory effects of pimozide on Kv currents. Pimozide induced membrane depolarization. We conclude that pimozide inhibits Kv currents in voltage-, time-, and use (state)-dependent manners. Furthermore, the major Kv channel target of pimozide is the Kv1.5 channel.


Assuntos
Antipsicóticos , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Animais , Coelhos , Antipsicóticos/toxicidade , Pimozida/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Músculo Liso Vascular , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Miócitos de Músculo Liso
10.
Seizure ; 101: 218-224, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36087422

RESUMO

Purpose The voltage-gated potassium channel Kv3.2, encoded by KCNC2, facilitates fast-spiking GABAergic interneurons to fire action potentials at high frequencies. It is pivotal to maintaining excitation/inhibition balance in mammalian brains. This study identified two novel de novo KCNC2 variants, p.Pro470Ser (P470S) and p.Phe382Leu (F382L), in patients with early onset developmental and epileptic encephalopathy (DEE). Methods To examine the molecular basis of DEE, we studied the functional characteristics of variant channels using patch-clamp techniques and computational modeling. Results Whole-cell patch clamp recordings from infected HEK293 cells revealed that channel activation and deactivation kinetics strongly decreased in both Kv3.2 P470S and F382L variant channels. This decrease also occurred in Kv3.2 p.Val471Leu (V471L) channels, known to be associated with DEE. In addition, Kv3.2 F382L and V471L variants exhibited a significant increase in channel conductance and a ∼20 mV negative shift in the threshold for voltage-dependent activation. Simulations of model GABAergic interneurons revealed that all variants decreased neuronal firing frequency. Thus, the variants' net loss-of-function effects disinhibited neural networks. Conclusion Our findings provide compelling evidence supporting the role of KCNC2 as a disease-causing gene in human neurodevelopmental delay and epilepsy.


Assuntos
Encefalopatias , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Potenciais de Ação/genética , Animais , Células HEK293 , Humanos , Mamíferos , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Canais de Potássio Shaw/genética
11.
Brain Stimul ; 15(3): 861-869, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35640845

RESUMO

BACKGROUND: Transcranial direct current stimulation (tDCS) is a non-invasive sub-threshold stimulation, widely accepted for its amelioration of distinct neuropsychiatric disorders. The weak electric field of tDCS modulates the activity of cortical neurons, which in turn modifies brain functioning. However, the underlying mechanisms for that are not fully understood. OBJECTIVE/HYPOTHESIS: Previous studies demonstrated that the axons are the most sensitive subcellular compartment for tDCS-induced polarization. Moreover, it was posited that DCS-induced axonal polarization is amplified by modifying the conductance of ionic channels. We posit that voltage-gated potassium-channels that are highly expressed in axons play a crucial role in DCS-induced modulation of cortical neurons functioning. METHODS: We examined the involvement of voltage-gated potassium-channels in the active modulation of spontaneous vesicle release by DCS. For that, we measured spontaneous excitatory postsynaptic currents (sEPSCs) from layer-V motor cortex during DCS application, while co-applying distinct voltage-gated potassium-channels blockers. Moreover, we examined the role of Kv1 potassium channels in DCS-induced modulation of action potential waveform at axon terminals by recording action potentials at terminal axon blebs during DCS application while locally inhibiting the Kv1 potassium-channels. RESULTS: We demonstrated that inhibiting voltage-gated potassium-channels occluded the DCS-induced modulation of subthreshold presynaptic vesicle release. Moreover, we showed that inhibiting Kv1 voltage-gated potassium-channels also occluded the DCS-induced modulation of action potential waveform at axon terminals. CONCLUSION: We suggest that DCS-induced depolarization inactivates the Kv1 potassium channels thus reducing potassium conductance, which amplifies axonal depolarization, subsequently enhancing the presynaptic component of synaptic transmission. Whereas DCS-induced hyperpolarization induces opposite effects.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Estimulação Transcraniana por Corrente Contínua , Potenciais de Ação/fisiologia , Axônios/fisiologia , Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Superfamília Shaker de Canais de Potássio/farmacologia
12.
Int J Mol Sci ; 22(3)2021 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-33572566

RESUMO

Ion channels are macromolecular complexes present in the plasma membrane and intracellular organelles of cells. Dysfunction of ion channels results in a group of disorders named channelopathies, which represent an extraordinary challenge for study and treatment. In this review, we will focus on voltage-gated potassium channels (KV), specifically on the KV4-family. The activation of these channels generates outward currents operating at subthreshold membrane potentials as recorded from myocardial cells (ITO, transient outward current) and from the somata of hippocampal neurons (ISA). In the heart, KV4 dysfunctions are related to Brugada syndrome, atrial fibrillation, hypertrophy, and heart failure. In hippocampus, KV4.x channelopathies are linked to schizophrenia, epilepsy, and Alzheimer's disease. KV4.x channels need to assemble with other accessory subunits (ß) to fully reproduce the ITO and ISA currents. ß Subunits affect channel gating and/or the traffic to the plasma membrane, and their dysfunctions may influence channel pharmacology. Among KV4 regulatory subunits, this review aims to analyze the KV4/KChIPs interaction and the effect of small molecule KChIP ligands in the A-type currents generated by the modulation of the KV4/KChIP channel complex. Knowledge gained from structural and functional studies using activators or inhibitors of the potassium current mediated by KV4/KChIPs will better help understand the underlying mechanism involving KV4-mediated-channelopathies, establishing the foundations for drug discovery, and hence their treatments.


Assuntos
Doença de Alzheimer/fisiopatologia , Canalopatias/fisiopatologia , Epilepsia/fisiopatologia , Proteínas Interatuantes com Canais de Kv/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Esquizofrenia/fisiopatologia , Canais de Potássio Shal/farmacologia , Doença de Alzheimer/etiologia , Sequência de Aminoácidos , Canalopatias/complicações , Epilepsia/etiologia , Coração/fisiopatologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Potenciais da Membrana , Modelos Moleculares , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Esquizofrenia/etiologia , Alinhamento de Sequência , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo
13.
Life Sci ; 91(11-12): 377-383, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22910181

RESUMO

AIMS: K(2P)2.1 (TREK1) two-pore-domain potassium channels control electrical activity in the central nervous system (CNS) and in the heart. Auxiliary ß subunits (Kvß) increase functional K+ channel diversity in the CNS. Based on similar tissue distribution and common functional significance of Kvß2 protein and K(2P)2.1 channels in neuronal excitability, we hypothesized that Kvß2 subunits modulate K2P2.1 currents. MAIN METHODS: Rat K2P2.1 channels and rKvß subunits were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to assess K2P2.1 function. KEY FINDINGS: Kvß2 subunits increased K(2P)2.1 currents by 2.9-fold in concentration-dependent fashion (I(0mV,K2P2.1), 0.53±0.07µA; I(0mV,K2P2.1+Kvß2), 1.56±0.13µA; n=15). K2P2.1 channel stimulation resulted in resting membrane potential hyperpolarization by -10.7mV (n=15). Open rectification and current-voltage relationships of K(2P)2.1 channels were not markedly altered upon co-expression with Kvß2, and K2P2.1 membrane expression was not affected by Kvß2 subunits. Related subunits Kvß1 (1.7-fold; n=16), Kvß3 (2.2-fold; n=16), and Kvß4 (2.8-fold; n=16) similarly activated K2P2.1 currents, indicating a broader role for Kvß proteins in K2P2.1 regulation. SIGNIFICANCE: Kvß subunits stabilize the resting membrane potential through enhancement of K2P2.1K+ currents. The significance of this previously unappreciated biophysical mechanism in neuronal physiology remains to be investigated.


Assuntos
Oócitos/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Western Blotting , Relação Dose-Resposta a Droga , Eletrodos , Eletrofisiologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Proteínas de Xenopus/farmacologia , Xenopus laevis
14.
Semin Perinatol ; 36(2): 92-7, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22414879

RESUMO

Closure of the ductus arteriosus at birth is a complex phenomenon being conditioned by antenatal events and progressing in preprogrammed steps. Functional at first, narrowing of the vessel is determined by 2 overlapping processes--removal of the prostaglandin E(2)-based relaxation sustaining prenatal patency and activation of a constrictor mechanism by the natural rise in blood oxygen tension. Two schemes have been proposed for oxygen action--one involving a cytochrome P450 hemoprotein (sensor)/endothelin-1 (effector) complex and the other a set of voltage-gated K(+) channels. These proposals, however, are not mutually exclusive. Structural closure follows the constriction through a remodeling process initiated antenatally with the development of intimal cushions and completed postnatally by a host of humoral and mechanical stimuli. Research in this area has already provided clinical applications. Nevertheless, management of premature infants with persistent ductus remains troublesome and calls for an alternative approach to the prostaglandin E(2) inhibitors now in use. Studies in progress on the oxygen-sensing system may lead to a definitive solution for this problem.


Assuntos
Permeabilidade do Canal Arterial/fisiopatologia , Doenças do Prematuro/fisiopatologia , Canal de Potássio Kv1.5/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/farmacologia , Espécies Reativas de Oxigênio/farmacologia , Remodelação Ventricular , Permeabilidade do Canal Arterial/tratamento farmacológico , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/tratamento farmacológico , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...