Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Ethnopharmacol ; 328: 117855, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38346524

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tea (Camellia sinensis) is a favorite drink worldwide. Tea extracts and green tea main component (-)-epigallocatechin gallate (EGCG) are recommended for various vascular diseases. Anji white tea is a very popular green tea. Its vascular effect profile, the mechanisms, and the contribution of EGCG to its integrated effect need elucidation. AIM: To characterize the vasomotion effects of Anji white tea and EGCG, and to explore possible involvement of voltage-gated Ca2+ channels (VGCCs) and voltage-gated K+ (Kv) channels in their vasomotion effects. MATERIALS AND METHODS: Anji white tea water soaking solution (AJWT) was prepared as daily tea-making process and concentrated to a concentration amounting to 200 mg/ml of dry tea leaves. The tension of rat arteries including aorta, coronary artery (RCA), cerebral basilar artery (CBA), intrarenal artery (IRA), intrapulmonary artery (IPA) and mesenteric artery (MA) was recorded with myographs. In arterial smooth muscle cells (ASMCs) freshly isolated from RCA, the levels of intracellular Ca2+ were measured with Ca2+-sensitive fluorescent probe fluo 4-AM, and Kv currents were recorded with patch clamp. The expressions of VGCCs and Kv channels were assayed with RT-qPCR and immunofluorescence staining. RESULTS: At 0.4-12.8 mg/ml of dry tea leaves, AJWT profoundly relaxed all tested arteries precontracted with various vasoconstrictors about half with a small transient potentiation on the precontractions before the relaxation. KCl-induced precontraction was less sensitive than precontractions induced by phenylephrine (PE), U46619 and serotonin (5-HT). IPA was less sensitive to the relaxation compared with other arteries. AJWT pretreatment for 1 h, 24 h and 72 h time-dependently inhibited the contractile responses of RCAs. In sharp contrast, at equivalent concentrations according to its content in AJWT, EGCG intensified the precontractions in most small arteries, except that it induced relaxation in PE-precontracted aorta and MA, U46619-precontracted aorta and CBA. EGCG pretreatment for 1 h and 24 h did not significantly affect RCA contractile responses. In RCA ASMCs, AJWT reduced, while EGCG enhanced, intracellular Ca2+ elevation induced by depolarization which activates VGCCs. Patch clamp study showed that both AJWT and EGCG reduced Kv currents. RT-qPCR and immunofluorescence staining demonstrated that both AJWT and EGCG reduced the expressions of VGCCs and Kv channels. CONCLUSION: AJWT, but not EGCG, consistently induces vasorelaxation. The vasomotion effects of either AJWT or EGCG vary with arterial beds and vasoconstrictors. Modulation of VGCCs, but not Kv channels, contributes to AJWT-induced vasorelaxation. It is suggested that Anji white tea water extract instead of EGCG may be a promising food supplement for vasospastic diseases.


Subject(s)
Catechin/analogs & derivatives , Myocytes, Smooth Muscle , Tea , Rats , Animals , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Vasodilation , Coronary Vessels , Mesenteric Arteries , Vasoconstrictor Agents/pharmacology , Water/pharmacology
2.
Eur J Pharmacol ; 957: 176005, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37611842

ABSTRACT

Lurasidone is a second-generation antipsychotic drug used to treat schizophrenia, mania, and bipolar disorder. The drug is an antagonist of the 5-HT2A and D2 receptors. No effect of lurasidone on the voltage-gated K+ (Kv) channels has yet been identified. Here, we show that lurasidone inhibits the vascular Kv channels of rabbit coronary arterial smooth muscle cells in a dose-dependent manner with an IC50 of 1.88 ± 0.21 µM and a Hill coefficient of 0.98 ± 0.09. Although lurasidone (3 µM) did not affect the activation kinetics, the drug negatively shifted the inactivation curve, suggesting that the drug interacted with the voltage sensors of Kv channels. Application of 1 or 2 Hz train steps in the presence of lurasidone significantly increased Kv current inhibition. The recovery time after channel inactivation increased in the presence of lurasidone. These results suggest that the inhibitory action of lurasidone is use (state)-dependent. Pretreatment with a Kv 1.5 subtype inhibitor effectively reduced the inhibitory effect of lurasidone. However, the inhibitory effect on Kv channels did not markedly change after pretreatment with a Kv 2.1 or a Kv7 subtype inhibitor. In summary, lurasidone inhibits vascular Kv channels (primarily the Kv1.5 subtype) in a concentration- and use (state)-dependent manner by shifting the steady-state inactivation curve.


Subject(s)
Antipsychotic Agents , Potassium Channels, Voltage-Gated , Animals , Rabbits , Lurasidone Hydrochloride/pharmacology , Antipsychotic Agents/pharmacology , Coronary Vessels , Myocytes, Smooth Muscle
3.
Annu Rev Biophys ; 52: 91-111, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36626766

ABSTRACT

Carefully orchestrated opening and closing of ion channels control the diffusion of ions across cell membranes, generating the electrical signals required for fast transmission of information throughout the nervous system. Inactivation is a parsimonious means for channels to restrict ion conduction without the need to remove the activating stimulus. Voltage-gated channel inactivation plays crucial physiological roles, such as controlling action potential duration and firing frequency in neurons. The ball-and-chain moniker applies to a type of inactivation proposed first for sodium channels and later shown to be a universal mechanism. Still, structural evidence for this mechanism remained elusive until recently. We review the ball-and-chain inactivation research starting from its introduction as a crucial component of sodium conductance during electrical signaling in the classical Hodgkin and Huxley studies, through the discovery of its simple intuitive mechanism in potassium channels during the molecular cloning era, to the eventual elucidation of a potassium channel structure in a ball-and-chain inactivated state.


Subject(s)
Potassium Channels , Signal Transduction , Potassium Channels/chemistry , Cell Membrane
4.
Clin Anat ; 36(2): 320-334, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36529666

ABSTRACT

The brain ventricular system (BVS) consists of brain ventricles and channels filled with cerebrospinal fluid (CSF). Disturbance of CSF flow has been linked to scoliosis and neurodegenerative diseases, including hydrocephalus. This could be due to defects of CSF production by the choroid plexus or impaired CSF movement over the ependyma dependent on motile cilia. Most vertebrates have horizontal body posture. They retain additional evolutionary innovations assisting CSF flow, such as the Reissner fiber. The causes of hydrocephalus have been studied using animal models including rodents (mice, rats, hamsters) and zebrafish. However, the horizontal body posture reduces the effect of gravity on CSF flow, which limits the use of mammalian models for scoliosis. In contrast, fish swim against the current and experience a forward-to-backward mechanical force akin to that caused by gravity in humans. This explains the increased popularity of the zebrafish model for studies of scoliosis. "Slit-ventricle" syndrome is another side of the spectrum of BVS anomalies. It develops because of insufficient inflation of the BVS. Recent advances in zebrafish functional genetics have revealed genes that could regulate the development of the BVS and CSF circulation. This review will describe the BVS of zebrafish, a typical teleost, and vertebrates in general, in comparative perspective. It will illustrate the usefulness of the zebrafish model for developmental studies of the choroid plexus (CP), CSF flow and the BVS.


Subject(s)
Hydrocephalus , Scoliosis , Humans , Cricetinae , Animals , Mice , Rats , Zebrafish/physiology , Cerebral Ventricles , Brain , Cerebrospinal Fluid/physiology , Mammals
5.
Mol Brain ; 15(1): 64, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35858950

ABSTRACT

Previous immunohistochemical studies have shown the expression of KCNQ2 channels at nodes of Ranvier (NRs) of myelinated nerves. However, functions of these channels at NRs remain elusive. In the present study, we addressed this issue by directly applying whole-cell patch-clamp recordings at NRs of rat lumbar spinal ventral nerves in ex vivo preparations. We show that depolarizing voltages evoke large non-inactivating outward currents at NRs, which are partially inhibited by KCNQ channel blocker linopirdine and potentiated by KCNQ channel activator retigabine. Furthermore, linopirdine significantly alters intrinsic electrophysiological properties of NRs to depolarize resting membrane potential, increase input resistance, prolong AP width, reduce AP threshold, and decrease AP amplitude. On the other hand, retigabine significantly decreases input resistance and increases AP rheobase at NRs. Moreover, linopirdine increases excitability at NRs by converting single AP firing into multiple AP firing at many NRs. Saltatory conduction velocity is significantly reduced by retigabine, and AP success rate at high stimulation frequency is significantly increased by linopirdine. Collectively, KCNQ2 channels play a significant role in regulating intrinsic electrophysiological properties and saltatory conduction at NRs of motor nerve fibers of rats. These findings may provide insights into how the loss-of-function mutation in KCNQ2 channels can lead to neuromuscular disorders in human patients.


Subject(s)
KCNQ2 Potassium Channel/metabolism , Ranvier's Nodes , Spinal Nerves , Animals , Electrophysiological Phenomena , KCNQ2 Potassium Channel/genetics , Membrane Potentials/physiology , Patch-Clamp Techniques , Ranvier's Nodes/metabolism , Rats
6.
Genes (Basel) ; 13(6)2022 06 20.
Article in English | MEDLINE | ID: mdl-35741862

ABSTRACT

Voltage-gated K+ channel ß subunits act as a structural component of Kin channels in different species. The ß subunits are not essential to the channel activity but confer different properties through binding the T1 domain or the C-terminal of α subunits. Here, we studied the physiological function of a novel gene, KIbB1, encoding a voltage-gated K+ channel ß subunit in sweetpotato. The transcriptional level of this gene was significantly higher in the low-K+-tolerant line than that in the low-K+-sensitive line under K+ deficiency conditions. In Arabidopsis, KIbB1 positively regulated low-K+ tolerance through regulating K+ uptake and translocation. Under high-salinity stress, the growth conditions of transgenic lines were obviously better than wild typr (WT). Enzymatic and non-enzymatic reactive oxygen species (ROS) scavenging were activated in transgenic plants. Accordingly, the malondialdehyde (MDA) content and the accumulation of ROS such as H2O2 and O2- were lower in transgenic lines under salt stress. It was also found that the overexpression of KIbB1 enhanced K+ uptake, but the translocation from root to shoot was not affected under salt stress. This demonstrates that KIbB1 acted as a positive regulator in high-salinity stress resistance through regulating Na+ and K+ uptake to maintain K+/Na+ homeostasis. These results collectively suggest that the mechanisms of KIbB1 in regulating K+ were somewhat different between low-K+ and high-salinity conditions.


Subject(s)
Arabidopsis , Ipomoea batatas , Homeostasis/genetics , Hydrogen Peroxide/metabolism , Ipomoea batatas/genetics , Reactive Oxygen Species/metabolism , Salt Tolerance/genetics
7.
J Neurosci ; 42(25): 4980-4994, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35606142

ABSTRACT

Ion channels at the nodes of Ranvier (NRs) are believed to play essential roles in intrinsic electrophysiological properties and saltatory conduction of action potentials (AP) at the NRs of myelinated nerves. While we have recently shown that two-pore domain potassium (K2P) channels play a key role at the NRs of Aß-afferent nerves, K+ channels and their functions at the NRs of mammalian motor nerves remain elusive. Here we addressed this issue by using ex vivo preparations of lumbar spinal ventral nerves from both male and female rats and the pressure-patch-clamp recordings at their NRs. We found that depolarizing voltages evoked large noninactivating outward currents at NRs. The outward currents could be partially inhibited by voltage-gated K+ channel blockers, largely inhibited by K2P blockers and cooling temperatures. Inhibition of the outward currents by voltage-gated K+ channel blockers, K2P blockers, or cooling temperatures significantly altered electrophysiological properties measured at the NRs, including resting membrane potential, input resistance, AP width, AP amplitude, AP threshold, and AP rheobase. Furthermore, K2P blockers and cooling temperatures significantly reduced saltatory conduction velocity and success rates of APs in response to high-frequency stimulation. Voltage-gated K+ channel blockers reduced AP success rates at high-frequency stimulation without significantly affecting saltatory conduction velocity. Collectively, both K2P and voltage-gated K+ channels play significant roles in intrinsic electrophysiological properties and saltatory conduction at NRs of motor nerve fibers of rats. The effects of cooling temperatures on saltatory conduction are at least partially mediated by K2P channels at the NRs.SIGNIFICANCE STATEMENT Ion channels localized at the NRs are believed to be key determinants of saltatory conduction on myelinated nerves. However, ion channels and their functions at the NRs have not been fully studied in different types of mammalian myelinated nerves. Here we use the pressure-patch-clamp recordings to show that both K2P and voltage-gated K+ channels play significant roles in intrinsic electrophysiological properties and saltatory conduction at NRs of lumbar spinal ventral nerves of rats. Furthermore, cooling temperatures exert effects on saltatory conduction via inhibition of ion channels at the NRs. Our results provide new insights into saltatory conduction on myelinated nerves and may have physiological as well as pathologic implications.


Subject(s)
Potassium Channels, Voltage-Gated , Action Potentials/physiology , Animals , Female , Male , Mammals , Membrane Potentials , Ranvier's Nodes , Rats , Spinal Nerves
8.
Exp Physiol ; 107(5): 441-449, 2022 05.
Article in English | MEDLINE | ID: mdl-35340063

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does inhibition of K+ channels modulate the exercise-training-induced augmentation in cholinergic and thermal sweating? What is the main finding and its importance? Iontophoretic administration of tetraethylammonium, a K+ channel blocker, blunted sweating induced by a low dose (0.001%) of the cholinergic agent pilocarpine, but not heat-induced sweating. However, no differences in the cholinergic sweating were observed between young endurance-trained and untrained men. Thus, while K+ channels play a role in the regulation of eccrine sweating, they do not contribute to the increase in sweating commonly observed in endurance-trained adults. Our findings provide important new insights into the mechanisms underlying the regulation of sweating by endurance conditioning. ABSTRACT: We evaluated the hypothesis that the activation of K+ channels mediates the exercise-training-induced augmentation of cholinergic and thermal sweating. On separate days, 11 endurance-trained and 10 untrained men participated in two experimental protocols. Prior to each protocol, we administered 2% tetraethylammonium (TEA, K+ channels blocker) and saline (Control) at forearm skin sites on both arms via transdermal iontophoresis. In protocol 1, low (0.001%) and high (1%) doses of pilocarpine were administered at the TEA-treated and Control sites over a 60-min period. In protocol 2, participants were passively heated by immersing their lower limbs in hot water (43°C) until core (rectal) temperature (Tc ) increased by 0.8°C above resting levels. Administration of TEA attenuated cholinergic sweating (P = 0.001) during the initial 20 min after the treatment of low dose of pilocarpine only whilst the response was similar between the groups (P = 0.163). Cholinergic and thermal sweating were higher in the trained relative to the untrained men (all P ≤ 0.033). Thermal sweating reached ∼90% of the response at a Tc elevation of 0.8°C during the initial 20 min of passive heating, which corresponds to the period wherein TEA attenuated cholinergic sweating in protocol 1. However, sweating did not differ between the Control and TEA sites in either group (P = 0.704). We showed that activation of K+ channels does not appear to mediate the elevated sweating response induced by a low dose of pilocarpine in trained men. We also demonstrated that K+ channels do not contribute to sweating during heat stress in either group.


Subject(s)
Endurance Training , Sweating , Adult , Cholinergic Agents , Humans , Male , Pilocarpine/pharmacology , Tetraethylammonium/pharmacology
9.
Clin Exp Pharmacol Physiol ; 48(5): 748-756, 2021 05.
Article in English | MEDLINE | ID: mdl-33620095

ABSTRACT

Tegaserod, a gastroprokinetic agent, is used to treat irritable bowel syndrome. Despite its extensive clinical use, little is known about the effects of tegaserod on vascular ion channels, especially K+ channels. Therefore, we examined the effects of tegaserod on voltage-gated K+ (Kv) channels in rabbit coronary arterial smooth muscle cells using the whole-cell patch-clamp technique. Tegaserod inhibited Kv channels in a concentration-dependent manner with an IC50 value of 1.26 ± 0.31 µmol/L and Hill coefficient of 0.81 ± 0.10. Although tegaserod had no effect on the steady-state activation curves of the Kv channels, the steady-state inactivation curve was shifted toward a more negative potential. These results suggest that tegaserod inhibits Kv channels by influencing their voltage sensors. The recovery time constant of channel inactivation was extended in the presence of tegaserod. Furthermore, application of train steps (1 and 2 Hz) in the presence of tegaserod progressively increased the inhibition of Kv currents suggesting that tegaserod-induced Kv channel inhibition is use (state)-dependent. Pretreatment with a Kv1.5 subtype inhibitor suppressed the Kv current. However, additional application of tegaserod did not induce further inhibition. Pretreatment with a Kv2.1 or Kv7 inhibitor did not affect the inhibitory effect of tegaserod on Kv channels. Based on these results, we conclude that tegaserod inhibits vascular Kv channels in a concentration- and use (state)-dependent manner independent of its own functions. Furthermore, the major Kv channel target of tegaserod is the Kv1.5 subtype.


Subject(s)
Indoles , Myocytes, Smooth Muscle , Animals , Muscle, Smooth, Vascular , Rabbits
10.
Endocrine ; 71(2): 365-377, 2021 02.
Article in English | MEDLINE | ID: mdl-33219494

ABSTRACT

PURPOSE: This study primarily investigated the effects of hypoglycemic compounds (Imeglimin derivatives) on insulin secretion in type 2 diabetes mellitus (T2DM), and further explored the possible mechanism underlying these effects. METHODS: Firstly, Metformin was used as the initiating compound to synthesize three sets of derivatives which contained Imeglimin structure core. At the cellular level, we screened compounds with better effect on the activity of insulin receptor tyrosine protein kinase (IFcTPK) after the islet ß cells were treated with the compounds of different concentrations. The insulin secretion was assessed using radioimmunoassay and the cytotoxicity to islet ß cells was evaluated by means of MTT assay following treatment with the compounds. The Ca2+-related mechanism by which these compounds promote insulin secretion was elucidated with whole cell recordings from current-clamp mode. RESULTS: Totally, 48 synthesized compounds were generated, wherein 10 compounds could increase the activity of IFcTPK in HIT-T15 cells better among these compounds. The modified Imeglimin, especially in the structure of hydrophilic hydroxyl or piperidine rings, could improve the activity of the compound to promote insulin secretion. Furthermore, the compounds 8a, 8b, 8k, and 9h revealed high insulin secretion-promoting activity. These compounds enhanced insulin secretion in islet ß cells by repressing the ATP-sensitive K(+) and voltage-gated K+ pathway. CONCLUSIONS: Our findings indicate that the hypoglycemic compounds 8a, 8b, 8k, and 9h confer better promotive effect on insulin secretion, which provides a reference for the development of drugs with better hypoglycemic activity.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Calcium/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism
11.
Eur J Pharmacol ; 891: 173707, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33137332

ABSTRACT

Darifenacin, an anticholinergic agent, has been used to treat overactive bladder syndrome. Despite its extensive clinical use, there is little information about the effect of darifenacin on vascular ion channels, specifically K+ channels. This study aimed to investigate the effect of the anti-muscarinic drug darifenacin on voltage-gated K+ (Kv) channels, vascular contractility, and coronary blood flow in rabbit coronary arteries. We used the whole-cell patch-clamp technique to evaluate the effect of darifenacin on Kv channels. Darifenacin inhibited the Kv current in a concentration-dependent manner. Applying 1 µM darifenacin shifted the activation and inactivation curves toward a more positive and negative potential, respectively. Darifenacin slowed the time constants of recovery from inactivation. Furthermore, blockade of the Kv current with darifenacin was increased gradually by applying a train of pulses, indicating that darifenacin inhibited Kv currents in a use- (state)-dependent manner. The darifenacin-mediated inhibition of Kv currents was associated with the Kv1.5 subtype, not the Kv2.1 or Kv7 subtype. Applying another anti-muscarinic drug atropine or ipratropium did not affect the Kv current or change the inhibitory effect of darifenacin. Isometric organ bath experiments using isolated coronary arteries were applied to evaluate whether darifenacin-induced inhibition of the Kv channel causes vasocontraction. Darifenacin substantially induced vasocontraction. Furthermore, darifenacin caused membrane depolarization and decreased coronary blood flow. From these results, we concluded that darifenacin inhibits the Kv currents in concentration- and use- (state)-dependent manners. Inhibition of the Kv current with darifenacin occurred by shifting the steady-state activation and inactivation curves regardless of its anti-muscarinic effect.


Subject(s)
Benzofurans/pharmacology , Coronary Vessels/drug effects , Kv1.5 Potassium Channel/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Potassium Channel Blockers/pharmacology , Pyrrolidines/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Kinetics , Kv1.5 Potassium Channel/metabolism , Male , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Rabbits
12.
BMC Cardiovasc Disord ; 20(1): 337, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32664860

ABSTRACT

BACKGROUND: High blood glucose impairs voltage-gated K+ (Kv) channel-mediated vasodilation in rat coronary artery smooth muscle cells (CSMCs) via oxidative stress. Advanced glycation end product (AGE) and receptor for AGE (RAGE) axis has been found to impair coronary dilation by reducing Kv channel activity in diabetic rat small coronary arteries (RSCAs). However, its underlying mechanism remain unclear. Here, we used isolated arteries and primary CSMCs to investigate the effect of AGE incubation on Kv channel-mediated coronary dilation and the possible involvement of peroxisome proliferators-activated receptor (PPAR) -γ pathway. METHODS: The RSCAs and primary CSMCs were isolated, cultured, and treated with bovine serum albumin (BSA), AGE-BSA, alagrebrium (ALA, AGE cross-linking breaker), pioglitazone (PIO, PPAR-γ activator) and/or GW9662 (PPAR-γ inhibitor). The groups were accordingly divided as control, BSA, AGE, AGE + ALA, AGE + PIO, or AGE + PIO + GW9662. Kv channel-mediated dilation was analyzed using wire myograph. Histology and immunohistochemistry of RSCAs were performed. Western blot was used to detect the protein expression of RAGE, major Kv channel subunits expressed in CSMCs (Kv1.2 and Kv1.5), PPAR-γ, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-2 (NOX-2). RESULTS: AGE markedly reduced Forskolin-induced Kv channel-mediated dilation of RSCAs by engaging with RAGE, and ALA or PIO significantly reversed the functional loss of Kv channel. In both RSCAs and CSMCs, AGE reduced Kv1.2/1.5 expression, increased RAGE and NOX-2 expression, and inhibited PPAR-γ expression, while ALA or PIO treatment partially reversed the inhibiting effects of AGE on Kv1.2/1.5 expression, accompanied by the downregulation of RAGE and decreased oxidative stress. Meanwhile, silencing of RAGE with siRNA remarkably alleviated the AGE-induced downregulation of Kv1.2/1.5 expression in CSMCs. CONCLUSION: AGE reduces the Kv channel expression in CSMCs and further impairs the Kv channel-mediated dilation in RSCAs. The AGE/RAGE axis may enhance oxidative stress by inhibiting the downstream PPAR-γ pathway, thus playing a critical role in the dysfunction of Kv channels.


Subject(s)
Glycation End Products, Advanced/pharmacology , Kv1.2 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , PPAR gamma/metabolism , Serum Albumin, Bovine/pharmacology , Vasodilation/drug effects , Anilides/pharmacology , Animals , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Kv1.2 Potassium Channel/genetics , Kv1.5 Potassium Channel/genetics , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , PPAR gamma/drug effects , Pioglitazone/pharmacology , Rats, Sprague-Dawley , Signal Transduction
13.
Biol Pharm Bull ; 43(7): 1123-1127, 2020.
Article in English | MEDLINE | ID: mdl-32612075

ABSTRACT

Nitric oxide (NO) is an important regulator of the retinal blood flow. The present study aimed to determine the role of voltage-gated K+ (KV) channels and ATP-sensitive K+ (KATP) channels in NO-mediated vasodilation of retinal arterioles in rats. In vivo, the retinal vasodilator responses were assessed by measuring changes in the diameter of retinal arterioles from ocular fundus images. Intravitreal injection of 4-aminopyridine (a KV channel inhibitor), but not glibenclamide (a KATP channel blocker), significantly attenuated the retinal vasodilator response to the NO donor (±)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide (NOR3). Intravitreal injection of indomethacin (a non-selective cyclooxygenase inhibitor) also reduced the NOR3-induced retinal vasodilator response. The combination of 4-aminopyridine and indomethacin produced a greater reduction in the NOR3-induced response than either agent alone. 4-Aminopyridine had no significant effect on pinacidil (a KATP channel opener)-induced response. These results suggest that the vasodilatory effects of NO are mediated, at least in part, through the activation of 4-aminopyridine-sensitive KV channels in the retinal arterioles of rats. NO exerts its dilatory effect on the retinal vasculature of rats through at least two mechanisms, activation of the KV channels and enhancement of prostaglandin production.


Subject(s)
4-Aminopyridine/pharmacology , Arterioles/drug effects , Nitric Oxide/physiology , Potassium Channel Blockers/pharmacology , Retinal Vessels/drug effects , Vasodilation/drug effects , Animals , Arterioles/physiology , Indomethacin/pharmacology , Male , Rats, Wistar , Retinal Vessels/physiology , Vasodilation/physiology
14.
Neuroscience ; 429: 256-263, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31962146

ABSTRACT

Myelinated Ah-type vagal ganglion neurons (VGNs) were specific subpopulation in adult females, rather than neonate and key players in sexual dimorphism in baroreflex afferent function and closely associated with estrogen. However, the gender related development changes in Ah-type VGNs remains unknown. To quantify the developmental changes in ion channels overtime, the whole-cell patch-clamp technique was performed and three afferent fiber types of VGNs were identified upon electrophysiological/pharmacological validations. The K+ currents were recorded with or without specific blockers from postnatal day 4-32 and adult in both sexes. The electrophysiological data conjugated with analysis of action potential (AP) trajectory strongly indicated that in male rats, Ah-types were likely to disappear or transform during development. The percentage of myelinated A-, Ah-, and unmyelinated C-type afferents in females remained relatively steady during the 4-32-day period. Conversely, Ah-type afferents in males declined from levels comparable with those in females at birth to near absence in adulthood at 32 days. The coordinated changes in the current density of certain ion channels may be the underlying mechanism of developmental changes in AP waveform and neuroexcitability. As expected, the coordinated change between the down-regulation of iberiotoxin-sensitive and up-regulation of 4-aminopyridine-sensitive K+ currents played a key role in shaping AP and neuroexcitability in Ah-types during development. Our results demonstrated that the myelinated Ah-type VGNs in males almost disappear at 4 weeks old where closes to adult and the correlative ion channel changes contribute to the sexual dimorphism in visceral afferent function.


Subject(s)
Neurons , Potassium Channels , Action Potentials , Animals , Female , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
15.
Cell Mol Life Sci ; 77(13): 2473-2482, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31894358

ABSTRACT

Voltage-gated K+ (Kv) channel opening repolarizes excitable cells by allowing K+ efflux. Over the last two decades, multiple Kv functions in the nervous system have been found to be unrelated to or beyond the immediate control of excitability, such as shaping action potential contours or regulation of inter-spike frequency. These functions include neuronal exocytosis and neurite formation, neuronal cell death, regulation of astrocyte Ca2+, glial cell and glioma proliferation. Some of these functions have been shown to be independent of K+ conduction, that is, they suggest the non-canonical functions of Kv channels. In this review, we focus on neuronal or glial plasmalemmal Kv channel functions which are unrelated to shaping action potentials or immediate control of excitability. Similar functions in other cell types will be discussed to some extent in appropriate contexts.


Subject(s)
Neuroglia/metabolism , Neurons/metabolism , Potassium Channels, Voltage-Gated/physiology , Action Potentials , Apoptosis , Astrocytes/metabolism , Calcium/metabolism , Cell Movement , Cell Proliferation , Exocytosis , Glioma/pathology , Neurites/physiology , Neuroglia/cytology
16.
Ecotoxicol Environ Saf ; 174: 58-65, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30822668

ABSTRACT

Di(2-ethylhexyl) phthalate (DEHP), as one of the most broadly representative phthalic acid esters, is used as a plasticizer in Polyvinyl chloride production. The exact neurotoxicologically effects of DEHP to human have not been adequately researched. In order to investigate the effects and mechanisms of DEHP exposure on neural circuit, the spatial learning and memory of Sprague Dawley (SD) rats was measured, and the cellular mechanisms underlying synaptic plasticity, cellular excitability and ion channels were detected. Our data showed that the spatial learning and memory was changed by DEHP (100 and 300 mg) treatment. Meanwhile, the frequency of mini Excitatory Postsynaptic Current (mEPSC) from CA3 pyramidal cells were significantly decreased by DEHP exposure (0.1 and 0.3 M); the firing threshold, membrane potential threshold, number, amplitude and latency of Action Potentials (Aps) of CA1 pyramidal cells were altered with the application of DEHP (0.1 and 0.3 M); furthermore, DEHP, both in 0.1 and 0.3 M could inhibit the voltage-gated potassium channel of CA1 pyramidal cells. Our results indicated that DEHP could impair the spatial learning and memory, and this impairment might due to the DEHP-induced suppression of the neuronal excitability and synaptic plasticity by inhibiting the voltage-gated potassium channel, supporting the hypothesis that DEHP could cause the disruption of neural function.


Subject(s)
Diethylhexyl Phthalate/toxicity , Memory/drug effects , Plasticizers/toxicity , Spatial Learning/drug effects , Animals , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley
17.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-30906529

ABSTRACT

Polyphosphoinositides (PPIn) are essential signaling phospholipids that make remarkable contributions to the identity of all cellular membranes and signaling cascades in mammalian cells. They exert regulatory control over membrane homeostasis via selective interactions with cellular proteins at the membrane-cytoplasm interface. This review article briefly summarizes our current understanding of the key roles that PPIn play in orchestrating and regulating crucial electrical and chemical signaling events in mammalian neurons and the significant neuro-pathophysiological conditions that arise following alterations in their metabolism.


Subject(s)
Neurons , Phosphatidylinositols , Signal Transduction , Animals , Cell Membrane , Neurons/physiology
18.
Eur J Pharmacol ; 849: 59-66, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30716319

ABSTRACT

In the present study, we investigated the inhibitory effect of tacrolimus, a macrolide immunosuppressive drug that acts through calcineurin inhibition, on voltage-gated K+ (Kv) channels expressed in native smooth muscle cells isolated from the coronary arteries of rabbits. Tacrolimus reduced the amplitude of Kv currents in a dose-dependent manner with an IC50 value and Hill coefficient of 7.80 ±â€¯3.01 µM and 1.07 ±â€¯0.25, respectively. Tacrolimus caused a shift in the activation curve toward a more positive potential and in the inactivation curve toward a more negative potential. Tacrolimus-induced inhibition of Kv current was increased by the application of train pulses (1 or 2 Hz). Furthermore, the recovery time constant of inactivation was extended in the presence of tacrolimus, suggesting that tacrolimus inhibited Kv channels in a use-dependent manner. Two kinds of Kv subtype inhibitors, DPO-1 and guangxitoxin did not affect the degree of tacrolimus-induced inhibition of Kv current. Furthermore, pretreatment with another calcineurin inhibitor, cyclosporine A, did not affect the Kv current, and did not alter the inhibitory effect of tacrolimus. Using perforated-patch clamp experiments, inhibition of Kv channels by tacrolimus caused membrane depolarization. From these results, we concluded that tacrolimus inhibited the vascular Kv currents in a dose, state (open and closed)-dependent manner.


Subject(s)
Coronary Vessels/cytology , Electrophysiological Phenomena/drug effects , Immunosuppressive Agents/pharmacology , KATP Channels/pharmacology , Myocytes, Smooth Muscle/drug effects , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Tacrolimus/pharmacology , Animals , Calcineurin/metabolism , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Male , Myocytes, Smooth Muscle/metabolism , Potassium Channels, Voltage-Gated/metabolism , Rabbits
19.
J Membr Biol ; 251(4): 573-579, 2018 08.
Article in English | MEDLINE | ID: mdl-29594433

ABSTRACT

Voltage-gated K+ (KV) currents play a crucial role in regulating pain by controlling neuronal excitability, and are divided into transient A-type currents (IA) and delayed rectifier currents (IK). The dorsal root ganglion (DRG) neurons are heterogeneous and the subtypes of KV currents display different levels in distinct cell sizes. To observe correlations of the subtypes of KV currents with DRG cell sizes, KV currents were recorded by whole-cell patch clamp in freshly isolated mouse DRG neurons. Results showed that IA occupied a high proportion in KV currents in medium- and large-diameter DRG neurons, whereas IK possessed a larger proportion of KV currents in small-diameter DRG neurons. A lower correlation was found between the proportion of IA or IK in KV currents and cell sizes. These data suggest that IA channels are mainly expressed in medium and large cells and IK channels are predominantly expressed in small cells.


Subject(s)
Ganglia, Spinal/metabolism , Potassium Channels, Voltage-Gated/metabolism , Animals , Ion Channel Gating/physiology , Membrane Potentials/physiology , Mice , Patch-Clamp Techniques
20.
Cell Mol Life Sci ; 75(3): 375-383, 2018 02.
Article in English | MEDLINE | ID: mdl-28780589

ABSTRACT

The brain ventricular system (BVS) consists of brain ventricles and channels connecting ventricles filled with cerebrospinal fluid (CSF). The disturbance of CSF flow has been linked to neurodegenerative disease including hydrocephalus, which manifests itself as an abnormal expansion of BVS. This relatively common developmental disorder has been observed in human and domesticated animals and linked to functional deficiency of various cells lineages facing BVS, including the choroid plexus or ependymal cells that generate CSF or the ciliated cells that cilia beating generates CSF flow. To understand the underlying causes of hydrocephalus, several animal models were developed, including rodents (mice, rat, and hamster) and zebrafish. At another side of a spectrum of BVS anomalies there is the "slit-ventricle" syndrome, which develops due to insufficient inflation of BVS. Recent advances in functional genetics of zebrafish brought to light novel genetic elements involved in development of BVS and circulation of CSF. This review aims to reveal common elements of morphologically different BVS of zebrafish as a typical representative of teleosts and other vertebrates and illustrate useful features of the zebrafish model for studies of BVS. Along this line, recent analyses of the two novel zebrafish mutants affecting different subunits of the potassium voltage-gated channels allowed to emphasize an important functional convergence of the evolutionarily conserved elements of protein transport essential for BVS development, which were revealed by the zebrafish and mouse studies.


Subject(s)
Cerebral Ventricles/embryology , Animals , Cerebral Ventricles/growth & development , Cricetinae , Humans , Mice , Rats , Vertebrates/embryology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...