1.GABA Receptor Activity Suppresses the Transition from Inter-ictal to Ictal Epileptiform Discharges in Juvenile Mouse Hippocampus.
Yan-Yan CHANG ; Xin-Wei GONG ; Hai-Qing GONG ; Pei-Ji LIANG ; Pu-Ming ZHANG ; Qin-Chi LU
Neuroscience Bulletin 2018;34(6):1007-1016
Exploring the transition from inter-ictal to ictal epileptiform discharges (IDs) and how GABA receptor-mediated action affects the onset of IDs will enrich our understanding of epileptogenesis and epilepsy treatment. We used Mg-free artificial cerebrospinal fluid (ACSF) to induce epileptiform discharges in juvenile mouse hippocampal slices and used a micro-electrode array to record the discharges. After the slices were exposed to Mg-free ACSF for 10 min-20 min, synchronous recurrent seizure-like events were recorded across the slices, and each event evolved from inter-ictal epileptiform discharges (IIDs) to pre-ictal epileptiform discharges (PIDs), and then to IDs. During the transition from IIDs to PIDs, the duration of discharges increased and the inter-discharge interval decreased. After adding 3 μmol/L of the GABA receptor agonist muscimol, PIDs and IDs disappeared, and IIDs remained. Further, the application of 10 μmol/L muscimol abolished all the epileptiform discharges. When the GABA receptor antagonist bicuculline was applied at 10 μmol/L, IIDs and PIDs disappeared, and IDs remained at decreased intervals. These results indicated that there are dynamic changes in the hippocampal network preceding the onset of IDs, and GABA receptor activity suppresses the transition from IIDs to IDs in juvenile mouse hippocampus.
Animals
;
Animals, Newborn
;
Bicuculline
;
pharmacology
;
Disease Models, Animal
;
Epilepsy
;
pathology
;
GABA-A Receptor Agonists
;
pharmacology
;
GABA-A Receptor Antagonists
;
therapeutic use
;
Hippocampus
;
drug effects
;
metabolism
;
physiopathology
;
In Vitro Techniques
;
Magnesium
;
metabolism
;
pharmacology
;
Male
;
Membrane Potentials
;
drug effects
;
Mice
;
Mice, Inbred C57BL
;
Muscimol
;
pharmacology
;
Nerve Net
;
drug effects
;
Receptors, GABA-A
;
metabolism
2.Cyproheptadine Regulates Pyramidal Neuron Excitability in Mouse Medial Prefrontal Cortex.
Yan-Lin HE ; Kai WANG ; Qian-Ru ZHAO ; Yan-Ai MEI
Neuroscience Bulletin 2018;34(5):759-768
Cyproheptadine (CPH), a first-generation antihistamine, enhances the delayed rectifier outward K current (I) in mouse cortical neurons through a sigma-1 receptor-mediated protein kinase A pathway. In this study, we aimed to determine the effects of CPH on neuronal excitability in current-clamped pyramidal neurons in mouse medial prefrontal cortex slices. CPH (10 µmol/L) significantly reduced the current density required to generate action potentials (APs) and increased the instantaneous frequency evoked by a depolarizing current. CPH also depolarized the resting membrane potential (RMP), decreased the delay time to elicit an AP, and reduced the spike threshold potential. This effect of CPH was mimicked by a sigma-1 receptor agonist and eliminated by an antagonist. Application of tetraethylammonium (TEA) to block I channels hyperpolarized the RMP and reduced the instantaneous frequency of APs. TEA eliminated the effects of CPH on AP frequency and delay time, but had no effect on spike threshold or RMP. The current-voltage relationship showed that CPH increased the membrane depolarization in response to positive current pulses and hyperpolarization in response to negative current pulses, suggesting that other types of membrane ion channels might also be affected by CPH. These results suggest that CPH increases the excitability of medial prefrontal cortex neurons by regulating TEA-sensitive I channels as well as other TEA-insensitive K channels, probably I and inward-rectifier Kir channels. This effect of CPH may explain its apparent clinical efficacy as an antidepressant and antipsychotic.
Animals
;
Cyproheptadine
;
pharmacology
;
Female
;
Histamine H1 Antagonists
;
pharmacology
;
Membrane Potentials
;
drug effects
;
physiology
;
Mice, Inbred C57BL
;
Patch-Clamp Techniques
;
Potassium Channel Blockers
;
pharmacology
;
Potassium Channels
;
metabolism
;
Prefrontal Cortex
;
drug effects
;
physiology
;
Pyramidal Cells
;
drug effects
;
physiology
;
Receptors, sigma
;
agonists
;
metabolism
;
Tetraethylammonium
;
pharmacology
;
Tissue Culture Techniques
3.The Role of Serotonin in Ventricular Repolarization in Pregnant Mice.
Shanyu CUI ; Hyewon PARK ; Hyelim PARK ; Dasom MUN ; Seung Hyun LEE ; Hyoeun KIM ; Nuri YUN ; Hail KIM ; Michael KIM ; Hui Nam PAK ; Moon Hyoung LEE ; Boyoung JOUNG
Yonsei Medical Journal 2018;59(2):279-286
PURPOSE: The mechanisms underlying repolarization abnormalities during pregnancy are not fully understood. Although maternal serotonin (5-hydroxytryptamine, 5-HT) production is an important determinant for normal fetal development in mice, its role in mothers remains unclear. We evaluated the role of serotonin in ventricular repolarization in mice hearts via 5Htr3 receptor (Htr3a) and investigated the mechanism of QT-prolongation during pregnancy. MATERIALS AND METHODS: We measured current amplitudes and the expression levels of voltage-gated K⁺ (Kv) channels in freshly-isolated left ventricular myocytes from wild-type non-pregnant (WT-NP), late-pregnant (WT-LP), and non-pregnant Htr3a homozygous knockout mice (Htr3a(−/−)-NP). RESULTS: During pregnancy, serotonin and tryptophan hydroxylase 1, a rate-limiting enzyme for the synthesis of serotonin, were markedly increased in hearts and serum. Serotonin increased Kv current densities concomitant with the shortening of the QT interval in WT-NP mice, but not in WT-LP and Htr3a(−/−)-NP mice. Ondansetron, an Htr3 antagonist, decreased Kv currents in WT-LP mice, but not in WT-NP mice. Kv4.3 directly interacted with Htr3a, and this binding was facilitated by serotonin. Serotonin increased the trafficking of Kv4.3 channels to the cellular membrane in WT-NP. CONCLUSION: Serotonin increases repolarizing currents by augmenting Kv currents. Elevated serotonin levels during pregnancy counterbalance pregnancy-related QT prolongation by facilitating Htr3-mediated Kv currents.
*Action Potentials/drug effects
;
Animals
;
Cell Membrane/drug effects/metabolism
;
Disease Models, Animal
;
Electrocardiography
;
Female
;
HSC70 Heat-Shock Proteins/metabolism
;
HSP90 Heat-Shock Proteins/metabolism
;
Heart Ventricles/drug effects/*metabolism
;
Mice, Inbred C57BL
;
Mice, Knockout
;
Myocytes, Cardiac/drug effects/metabolism
;
Potassium Channels/metabolism
;
Pregnancy
;
Rabbits
;
Rats, Sprague-Dawley
;
Receptors, Serotonin, 5-HT3/metabolism
;
Serotonin/*metabolism
;
Serotonin 5-HT3 Receptor Agonists/pharmacology
4.Antiarrhythmic ionic mechanism of Guanfu base A--Selective inhibition of late sodium current in isolated ventricular myocytes from guinea pigs.
Si-Si JIN ; Qiao GUO ; Jing XU ; Peng YU ; Jing-Han LIU ; Yi-Qun TANG
Chinese Journal of Natural Medicines (English Ed.) 2015;13(5):361-367
The present study was designed to determine the effects of Guanfu base A (GFA) on the late sodium current (INa.L), transient sodium current (INa.T), HERG current (IHERG), and Kv1.5 current (IKv1.5). The values of INa.L, INa.T, IHERG and IKv1.5 were recorded using the whole-cell patch clamp technique. Compared with other channels, GFA showed selective blocking activity in late sodium channel. It inhibited INa.L in a concentration-dependent manner with an IC50 of (1.57 ± 0.14) μmol · L(-1), which was significantly lower than its IC50 values of (21.17 ± 4.51) μmol · L(-1) for the INa.T. The inhibitory effect of GFA on INa,L was not affected by 200 μmol · L(-1) H2O2. It inhibited IHERG with an IC50 of (273 ± 34) μmol · L(-1) and has slight blocking effect on IKv1.5, decreasing IKv1.5 by only 20.6% at 200 μmol · L(-1). In summary, GFA inhibited INa.L selectively and remained similar inhibition in presence of reactive oxygen species. These findings may suggest a novel molecular mechanism for the potential clinical application of GFA in the treatment of cardiovascular disorders.
Analysis of Variance
;
Animals
;
Anti-Arrhythmia Agents
;
pharmacology
;
Dose-Response Relationship, Drug
;
Female
;
Guinea Pigs
;
HEK293 Cells
;
Heart Ventricles
;
drug effects
;
Heterocyclic Compounds, 4 or More Rings
;
pharmacology
;
Humans
;
Inhibitory Concentration 50
;
Male
;
Membrane Potentials
;
drug effects
;
Myocytes, Cardiac
;
metabolism
;
Patch-Clamp Techniques
;
Sodium Channel Blockers
;
pharmacology
;
Sodium Channels
;
drug effects
5.Pacilitaxel induces human nasopharyngeal carcinoma cell line CNE2 apoptosis and growth inhibition by suppressing PI3K/AKT/p53 signaling pathway.
Journal of Clinical Otorhinolaryngology Head and Neck Surgery 2015;29(24):2147-2150
OBJECTIVE:
To investigate the effect and mechanisms of the PTX on the human nasopharyngeal carcinoma cell line CNE2.
METHOD:
Cells from CNE2 were cultured in vitro and the cells at logarithmic growth phase were processed with different concentration of PTX (0, 5, 10, 20) mol/L for 72h. MTT was used to evaluate the proliferation and flow cytometric analysis was utilized to detect membrane potential and apoptosis of CNE2 cells. The expression of PI3K, p-AKT, AKT, p53, p21, Caspase3, Cleavage-Caspase3, PARP, Cleavage-PARP, AIF, CytC, Bcl-2 and Bax in CNE2 cells were examined by Western Blot.
RESULT:
The results showed that PTX could increase the apoptosis and the expression of Caspase3, PARP, CytC, AIF and Bax and reduce the proliferation, membrane potential and the expression of PI3K, p-AKT, p53, p21, Cleavage-PARP, Cleavage-Caspase3 and Bcl-2 in CNE2 cell in a concentration-dependent manner. However, PTX had no effect on the expression of AKT.
CONCLUSION
PTX can promote apoptosis and growth inhibition of human nasopharyngeal cancer cell line CNE2 and the mechanism involves suppressing PI3K/AKT/p53 signaling pathway.
Apoptosis
;
Apoptosis Regulatory Proteins
;
metabolism
;
Carcinoma
;
Cell Line, Tumor
;
drug effects
;
Flow Cytometry
;
Humans
;
Membrane Potentials
;
Nasopharyngeal Carcinoma
;
Nasopharyngeal Neoplasms
;
metabolism
;
pathology
;
Paclitaxel
;
pharmacology
;
Phosphatidylinositol 3-Kinases
;
metabolism
;
Proto-Oncogene Proteins c-akt
;
metabolism
;
Signal Transduction
;
Tumor Suppressor Protein p53
;
metabolism
6.Effect of hydrogen peroxide on electric current of large-conductance calcium-activated potassium channel in isolated outer hair cells of old guinea pig cochlea.
Acta Physiologica Sinica 2014;66(3):302-306
The present study was aimed to investigate the effect of hydrogen peroxide (H₂O₂, oxygen free radical donator) on the current of large-conductance calcium-activated potassium channels (BK(Ca) channels) in isolated outer hair cells of old guinea pig cochlea, and to explore the underlying mechanism. Outer hair cells of old guinea pig cochlea were acutely enzyme-isolated, and currents were recorded by whole-cell patch clamp. The results showed that, rapid activation and non-deactivation electric currents with a string of large amplitude were recorded. Activation voltage of the current was above -40 - -30 mV. The amplitude of current was increased continuously with the rising of membrane potential. The current showed characteristics of outward rectification without "rundown" phenomenon. IbTX (100 nmol/L) could completely block the activity of channel, which confirmed BK(Ca) channel's current. BK(Ca) current amplitude and peak current density increased with the increment of H₂O₂ concentration (1, 2, 4 μmol/L), showing concentration-dependent activation by H₂O₂. Our results suggest that oxygen free radical/BK(Ca) pathway may be able to adjust the balance of intracellular calcium in outer hair cells.
Animals
;
Calcium
;
metabolism
;
Cochlea
;
cytology
;
Guinea Pigs
;
Hair Cells, Vestibular
;
drug effects
;
Hydrogen Peroxide
;
pharmacology
;
Large-Conductance Calcium-Activated Potassium Channels
;
metabolism
;
Membrane Potentials
7.Chlorogenic acid alters the voltage-gated potassium channel currents of trigeminal ganglion neurons.
Yu-Jiao ZHANG ; Xiao-Wen LU ; Ning SONG ; Liang KOU ; Min-Ke WU ; Fei LIU ; Hang WANG ; Jie-Fei SHEN
International Journal of Oral Science 2014;6(4):233-240
Chlorogenic acid (5-caffeoylquinic acid, CGA) is a phenolic compound that is found ubiquitously in plants, fruits and vegetables and is formed via the esterification of caffeic acid and quinic acid. In addition to its notable biological functions against cardiovascular diseases, type-2 diabetes and inflammatory conditions, CGA was recently hypothesized to be an alternative for the treatment of neurological diseases such as Alzheimer's disease and neuropathic pain disorders. However, its mechanism of action is unclear. Voltage-gated potassium channel (Kv) is a crucial factor in the electro-physiological processes of sensory neurons. Kv has also been identified as a potential therapeutic target for inflammation and neuropathic pain disorders. In this study, we analysed the effects of CGA on the two main subtypes of Kv in trigeminal ganglion neurons, namely, the IK,A and IK,V channels. Trigeminal ganglion (TRG) neurons were acutely disassociated from the rat TRG, and two different doses of CGA (0.2 and 1 mmol⋅L(-1)) were applied to the cells. Whole-cell patch-clamp recordings were performed to observe alterations in the activation and inactivation properties of the IK,A and IK,V channels. The results demonstrated that 0.2 mmol⋅L(-1) CGA decreased the peak current density of IK,A. Both 0.2 mmol⋅L(-1) and 1 mmol⋅L(-1) CGA also caused a significant reduction in the activation and inactivation thresholds of IK,A and IK,V. CGA exhibited a strong effect on the activation and inactivation velocities of IK,A and IK,V. These findings provide novel evidence explaining the biological effects of CGA, especially regarding its neurological effects.
Analgesics
;
pharmacology
;
Animals
;
Animals, Newborn
;
Cell Culture Techniques
;
Chlorogenic Acid
;
administration & dosage
;
pharmacology
;
Ion Channel Gating
;
drug effects
;
Membrane Potentials
;
drug effects
;
Neurons
;
drug effects
;
Neurotransmitter Agents
;
administration & dosage
;
pharmacology
;
Patch-Clamp Techniques
;
Potassium Channels, Voltage-Gated
;
drug effects
;
Rats
;
Rats, Sprague-Dawley
;
Trigeminal Ganglion
;
drug effects
8.Enhancement of GABA-activated currents by arginine vasopressin in rat dorsal root ganglion neurons.
Fang QIU ; Wang-Ping HU ; Zhi-Fan YANG
Acta Physiologica Sinica 2014;66(6):647-657
A growing number of studies have shown that arginine vasopressin (AVP) plays an analgesia role in the modulation of nociception. Previous studies have focused on the central mechanisms of AVP analgesia. The aim of the present study was to find out whether peripheral mechanisms are also involved. The effect of AVP on GABA-activated currents (IGABA) and GABAA receptor function in freshly isolated dorsal root ganglion (DRG) neurons of rats were studied using whole cell patch clamp technique. The result showed that, IGABA were potentiated by pre-treatment with AVP (1 × 10⁻¹⁰-1 × 10⁻⁵ mol/L) in a concentration-dependent manner. Meanwhile, the GABA concentration-response curve was shifted upwards, with an increase of (49.1 ± 4.0)% in the maximal current response but with no significant change in the EC50 values. These results indicate that the enhancing effect is non-competitive. In addition, the effects of AVP on IGABA might be voltage-independent. This potentiation of IGABA induced by AVP was almost completely blocked by the V1a receptor antagonist SR49059 (3 × 10⁻⁶ mol/L). Also it could be removed by intracellular dialysis of either GDP-β-S (5 × 10⁻⁴mol/L), a non-hydrolyzable GDP analog, or GF109203X (2 × 10⁻⁶ mol/L), a selective protein kinase C (PKC) inhibitor, with the re-patch clamp. These results suggest that AVP up-regulates the function of the GABAA receptor via G protein-coupled receptors and PKC-dependent signal pathways in rat DRG neurons, and this potentiation may underlie the analgesia induced by AVP.
Animals
;
Arginine Vasopressin
;
pharmacology
;
Ganglia, Spinal
;
cytology
;
Guanosine Diphosphate
;
analogs & derivatives
;
pharmacology
;
Indoles
;
Maleimides
;
Membrane Potentials
;
Neurons
;
drug effects
;
Patch-Clamp Techniques
;
Rats
;
Rats, Sprague-Dawley
;
Receptors, GABA-A
;
metabolism
;
Signal Transduction
;
Thionucleotides
;
pharmacology
;
gamma-Aminobutyric Acid
;
pharmacology
9.The effect of niflumic acid and blocker of calcium channel on the desensitization of gamma aminobutyric acid-activated current.
Li LI ; Yang WANG ; Ke-Tao MA ; Hong-Ju CHENG ; Lei ZHAO ; Jun-Qiang SI
Chinese Journal of Applied Physiology 2013;29(2):128-132
OBJECTIVETo explore the modulatory effect of niflumic acid and blocker of calcium channel on the desensitization of gamma aminobutyric acid (GABA)-activated currents in dorsal root ganglion(DRG) neurons from rat.
METHODSThe whole-cell patch-clamp technique was used to observe the modulatory effect of niflumic acid and blocker of calcium channel on the desensitization of GABA-activated currents in neurons freshly dissociated from rat DRG neurons.
RESULTSApplication of GABA (0.1-1 000 micromol/L) could induce concentration-dependent inward currents in some cells (212/223, 95.11%). GABA-(100 micromol/L) activated currents was (1.32 +/- 0.74) nA (n = 84). However, pre-application of niflumic acid (1-100 micromol/L) and nitrendipine (specific blocker of L-calcium channel)(0.1-30 micromol/L) could inhibit the GABA-activated inward current which was identified to be GABAA receptor-mediated current. The inhibitory effects of niflumic acid and nitrendipine were concentration-dependent. The suppression rate of 10 micromol/L niflumic acid and nitrendipine to GABA-activated currents were (31.60% +/- 4.87%) (n = 19) and (43.60% < or = 5.10%) (n = 5), respectively. The desensitization of GABA-activated currents had double exponential characteristic. Tau value was (14.68 +/- 5.11) s (n = 6) and (175.8 +/- 42.67) s (n = 6, r = 0.9647), respectively. Pre-application of niflumic acid (100 micromol/L) and nickel chloride (nonspecific blocker of L-calcium channel) (100 micromol/L) altered tau value of the desensitization of GABA-activated currents, tau value reduced for (4.64 +/- 2.21) s (n = 3), (43.70 +/- 14.34) s ( n = 3, r = 0.9548) and (4.64 +/- 2.21) s (n = 3), (43.70 +/- 14.34) s (n = 3, r = 0.9721).
CONCLUSIONPre-application of niflumic acid exerts a more strong inhibitory effect on the peak value of GABA-activated current, which possibly is through blocking the calcium-activated chloride ion channel to accelerate the desensitization of GABA-activated currents.
Animals ; Animals, Newborn ; Calcium Channel Blockers ; pharmacology ; Calcium Channels, L-Type ; drug effects ; Ganglia, Spinal ; drug effects ; physiology ; Membrane Potentials ; drug effects ; physiology ; Neurons ; drug effects ; physiology ; Niflumic Acid ; pharmacology ; Nitrendipine ; pharmacology ; Patch-Clamp Techniques ; Rats ; Rats, Sprague-Dawley ; gamma-Aminobutyric Acid ; pharmacology
10.Effects of ropivacaine on GABA-activated currents in isolated dorsal root ganglion neurons in rats.
Yue YANG ; Jun-Qiang SI ; Chao FAN ; Ke-Tao MA ; Hong-Jv CHENG ; Li LI
Chinese Journal of Applied Physiology 2013;29(3):263-266
OBJECTIVETo investigate the effects of ropivacaine on Gamma-aminobutyric acid(GABA)-activated currents in dorsal root ganglion (DRG) neurons in rats and discuss the analgesia mechanism of ropivacaine.
METHODSBy means of using whole-cell patch-clamp technique, to investigate the modulatory effects of ropivacaine on GABA-activated currents (I(GABA)) in acutely isolated dorsal root ganglion neurons.
RESULTS(1) In 48 out of 73DRG cells (65.7%, 48/73), to perfusion ropivacaine bromide (0.1 - 1 000 micromol/L) were sensitive. Which produce in 0 to 380 pA current. (2) The majority of the neurons examined (74.5%, 73/98) were sensitive to GABA. Concentration of 1 - 1 000 micromol/L GABA could activate a concentration-dependent inward current, which manifested obvious desensitization, and the inward currents could be blocked byGABA-receptor selective antagonist of bicuculline (100 micromol/L). (3) After the neurons were treated with ropivacaine (0.1 - 1000 micromol/L) prior to the application of GABA (100 micromol/L) 30 s, GABA currents were obviously increased. Ropivacaine could make dose-response curve of the GABA up, EC50 is 23.46 micromol/L. Ropivacaine shifted the GABA dose-response curve upward and increased the maximum response to the contrast about 153%.
CONCLUSIONThe enhancement of ropivacaine to DRG neurons activation of GABA current, can lead to enhancement of pre-synaptic inhibition at the spinal cord level. This may be one of the reasons for the anesthetic effect and analgesia for ropivacaine in epidural anesthesia.
Amides ; pharmacology ; Animals ; Ganglia, Spinal ; cytology ; physiology ; Membrane Potentials ; drug effects ; Neurons ; cytology ; drug effects ; physiology ; Patch-Clamp Techniques ; Rats ; Rats, Sprague-Dawley ; Receptors, GABA-A ; physiology

Result Analysis
Print
Save
E-mail