Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Mol Neurosci ; 72(12): 2464-2472, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36508141

ABSTRACT

This study was designed to determine the effects and underlying mechanism of honokiol (HNK) on traumatic brain injury (TBI). A rat TBI model was constructed using the modified Feeney free-fall percussion method and treatment with HNK via intraperitoneal injection. The brain tissues of the rats in each group were assessed using the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay to detect the level of neuronal apoptosis. Western blots were used to detect the expression levels of apoptosis-related proteins (Bcl-2 and Bax), and ELISAs were used to measure the levels of pro-inflammatory cytokines (IL-18 and IL-1ß) and the activity of caspase-1. In addition, the mitochondrial membrane potential, reactive oxygen species (ROS), and adenosine 5'-triphosphate (ATP) were also measured. Western blots and qRT-PCRs were used to determine the relative expression levels of the mitochondrial unfolded protein response (UPRmt)-related proteins and mRNAs. Based on the experimental results, treatment with HNK was associated with a decrease in the number of TUNEL-positive cells, downregulated Bax expression levels, elevated Bcl-2 expression levels, and inhibition of neuronal apoptosis in the brain tissue of TBI rats. HNK also suppressed neuroinflammation by decreasing IL-1ß and IL-18 levels and caspase-1 activity. Additionally, HNK lowered the mitochondrial membrane potential and ROS levels, increased ATP levels, and improved mitochondrial dysfunction in neural cells. Furthermore, in the investigation of the mechanism of HNK on TBI, we observed that HNK could activate UPRmt by upregulating the mRNA and protein expression levels of HSPA9, CLPP, and HSP60 in the brain tissues of TBI rats. Collectively, HNK reduced mitochondrial dysfunction, inhibited the apoptosis of nerve cells, and attenuated inflammation in the brains of TBI rats. The protective effect of HNK may be achieved through the activation of UPRmt.


Subject(s)
Brain Injuries, Traumatic , Interleukin-18 , Rats , Animals , Interleukin-18/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Neurons/metabolism , Unfolded Protein Response , Mitochondria/metabolism , Caspases/metabolism , Caspases/pharmacology
2.
Chin Med J (Engl) ; 129(6): 651-6, 2016 Mar 20.
Article in English | MEDLINE | ID: mdl-26960367

ABSTRACT

BACKGROUND: Obstructive sleep apnea (OSA) is a common sleep disorder and is characterized by airway collapse at multiple levels of upper airway. The effectiveness of nasal surgery has been discussed in several studies and shows a promising growing interest. In this study, we intended to evaluate the effects of nasal surgery on the upper airway dimensions in patients with OSA using three-dimensional (3D) reconstruction of cone-beam computed tomography (CT). METHODS: Twelve patients with moderate to severe OSA who underwent nasal surgery were included in this study. All patients were diagnosed with OSA using polysomnography (PSG) in multi sleep health centers associated with Massachusetts General Hospital, Massachusetts Eye and Ear Infirmary and the Partners Health Care from May 31, 2011 to December 14, 2013. The effect of nasal surgery was evaluated by the examination of PSG, subjective complains, and 3D reconstructed CT scan. Cross-sectional area was measured in eleven coronal levels, and nasal cavity volume was evaluated from anterior nasal spine to posterior nasal spine. The thickness of soft tissue in oral pharynx region was also measured. RESULTS: Five out of the 12 patients were successfully treated by nasal surgery, with more than 50% drop of apnea-hypopnea index. All the 12 patients showed significant increase of cross-sectional area and volume postoperatively. The thickness of soft tissue in oral pharynx region revealed significant decrease postoperatively, which decreased from 19.14 ± 2.40 cm 2 and 6.11 ± 1.76 cm 2 to 17.13 ± 1.91 cm 2 and 5.22 ± 1.20 cm 2 . CONCLUSIONS: Nasal surgery improved OSA severity as measured by PSG, subjective complaints, and 3D reconstructed CT scan. 3D assessment of upper airway can play an important role in the evaluation of treatment outcome.


Subject(s)
Cone-Beam Computed Tomography/methods , Imaging, Three-Dimensional/methods , Nasal Surgical Procedures , Sleep Apnea, Obstructive/surgery , Adult , Female , Humans , Male , Middle Aged , Quality of Life , Sleep Apnea, Obstructive/diagnostic imaging , Sleep Apnea, Obstructive/psychology
3.
Cell Physiol Biochem ; 36(5): 1699-711, 2015.
Article in English | MEDLINE | ID: mdl-26183394

ABSTRACT

AIMS: PGE2 is one of the most abundant prostanoids in mammalian tissues, but its effect on neuronal receptors has not been well investigated. This study examines the effect of PGE2 on GABAA receptor currents in rat cerebellar granule neurons. METHODS: GABAA currents were recorded using a patch-clamp technique. Cell surface and total protein of GABAA ß1/2/3 subunits was carried out by Western blot analysis. RESULTS: Upon incubation of neurons with PGE2 (1 µM) for 60 minutes, GABAA currents were significantly potentiated. This PGE2-driven effect could be blocked by PKC or CaMKII inhibitors as well as EP1 receptor antagonist, and mimicked by PMA or EP1 receptor agonist. Furthermore, Western blot data showed that PGE2 did not increase the total expression level of GABAA receptors, but significantly increased surface levels of GABAA ß1/2/3 subunits after 1 h of treatment. Consistently, both PKC and CaMKII inhibitors were able to reduce PGE2-induced increases in cell surface expression of GABAA receptors. CONCLUSION: Activation of either the PKC or CaMKII pathways by EP1 receptors mediates the PGE2-induced increase in GABAA currents. This suggests that upregulation of postsynaptic GABAA receptors by PGE2 may have profound effects on cerebellar functioning under physiological and pathological conditions.


Subject(s)
Dinoprostone/physiology , Receptors, GABA-A/physiology , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Signal Transduction , Animals , Cells, Cultured , Cerebellum/metabolism , Cytoplasmic Granules/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
4.
J Biol Chem ; 287(49): 41534-45, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23066017

ABSTRACT

Neuritin is a new neurotrophic factor discovered in a screen to identify genes involved in activity-dependent synaptic plasticity. Neuritin also plays multiple roles in the process of neural development and synaptic plasticity. The receptors for binding neuritin and its downstream signaling effectors, however, remain unclear. Here, we report that neuritin specifically increases the densities of transient outward K(+) currents (I(A)) in rat cerebellar granule neurons (CGNs) in a time- and concentration-dependent manner. Neuritin-induced amplification of I(A) is mediated by increased mRNA and protein expression of Kv4.2, the main α-subunit of I(A). Exposure of CGNs to neuritin markedly induces phosphorylation of ERK (pERK), Akt (pAkt), and mammalian target of rapamycin (pmTOR). Neuritin-induced I(A) and increased expression of Kv4.2 are attenuated by ERK, Akt, or mTOR inhibitors. Unexpectedly, pharmacological blockade of insulin receptor, but not the insulin-like growth factor 1 receptor, abrogates the effect of neuritin on I(A) amplification and Kv4.2 induction. Indeed, neuritin activates downstream signaling effectors of the insulin receptor in CGNs and HeLa. Our data reveal, for the first time, an unanticipated role of the insulin receptor in previously unrecognized neuritin-mediated signaling.


Subject(s)
Cerebellum/metabolism , Gene Expression Regulation , Neurons/metabolism , Neuropeptides/metabolism , Receptor, Insulin/metabolism , Shal Potassium Channels/metabolism , Animals , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , GPI-Linked Proteins/metabolism , HeLa Cells , Humans , Kinetics , Models, Biological , Patch-Clamp Techniques , Rats , Up-Regulation
5.
PLoS One ; 7(7): e41303, 2012.
Article in English | MEDLINE | ID: mdl-22844454

ABSTRACT

Cyproheptadine (CPH) is a histamine- and serotonin-receptor antagonist, and its effects are observed recently in the modulation of multiple intracellular signals. In this study, we used cortical neurons and HEK-293 cells transfected with Kv2.1 α-subunit to address whether CPH modify neural voltage-gated K(+) channels by a mechanism independent of its serotonergic and histaminergic properties. Our results demonstrate that intracellularly delivered CPH increased the I(K) by reducing the activity of protein kinas A (PKA). Inhibition of G(i) eliminated the CPH-induced effect on both the I(K) and PKA. Blocking of 5-HT-, M-, D(2)-, H(1)- or H(2)-type GPCR receptors with relevant antagonists did not eliminate the CPH-induced effect on the I(K). Antagonists of the sigma-1 receptor, however, blocked the effect of CPH. Moreover, the inhibition of sigma-1 by siRNA knockdown significantly reduced the CPH-induced effect on the I(K). On the contrary, sigma-1 receptor agonist mimicked the effects of CPH on the induction of I(K). A ligand-receptor binding assay indicated that CPH bound to the sigma-1 receptor. Similar effect of CPH were obtained from HEK-293 cells transfected with the α-subunit of Kv2.1. In overall, we reveal for the first time that CPH enhances the I(K) by modulating activity of PKA, and that the associated activation of the sigma-1 receptor/G(i)-protein pathway might be involved. Our findings illustrate an uncharacterized effect of CPH on neuron excitability through the I(K), which is independent of histamine H(1) and serotonin receptors.


Subject(s)
Cerebral Cortex/cytology , Cyproheptadine/pharmacology , Intracellular Space/drug effects , Neurons/drug effects , Potassium/metabolism , Receptors, sigma/metabolism , Signal Transduction/drug effects , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Electric Conductivity , Extracellular Space/drug effects , Extracellular Space/metabolism , HEK293 Cells , Histamine Antagonists/pharmacology , Humans , Intracellular Space/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Shab Potassium Channels/metabolism , Sigma-1 Receptor
6.
J Neurochem ; 120(4): 502-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22118516

ABSTRACT

Cholesterol is a major component of membrane lipid rafts. It is more abundant in the brain than in other tissues and plays a critical role in maintaining brain function. We report here that a significant enhancement in apoptosis in rat cerebellar granule neurons (CGNs) was observed upon incubation with 5mM K(+) /serum free (LK-S) medium. Cholesterol enrichment further potentiated CGN apoptosis incubated under LK-S medium. On the contrary, cholesterol depletion using methyl-beta-cyclodextrin protected the CGNs from apoptosis induced by LK-S treatment. Cholesterol enrichment, however, did not induce apoptosis in CGNs that have been incubated with 25mM K(+) /serum medium. Mechanistically, increased I(K) currents and DNA fragmentation were found in CGNs incubated in LK-S, which was further potentiated in the presence of cholesterol. Cholesterol-treated CGNs also exhibited increased cAMP levels and up-regulation of Kv2.1 expression. Increased levels of activated form of PKA and phospho-CREB further supported activation of the cAMP/PKA pathway upon treatment of CGNs with cholesterol-containing LK-S medium. Conversely, inhibition of PKA or small G protein Gs abolished the increase in I(K) current and the potentiation of Kv2.1 expression, leading to reduced susceptibility of CGNs to LK-S and cholesterol-induced apoptosis. Our results demonstrate that the elevation of membrane cholesterol enhances CGN susceptibility to apoptotic stimuli via cAMP/PKA/CREB-dependent up-regulation of Kv2.1. Our data provide new evidence for the role of cholesterol in eliciting neuronal cell death.


Subject(s)
Apoptosis/genetics , CREB-Binding Protein/metabolism , Cholesterol/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Neurons/metabolism , Shab Potassium Channels/biosynthesis , Up-Regulation/physiology , Animals , Cells, Cultured , Cyclic AMP/physiology , Female , Genetic Predisposition to Disease , Rats , Rats, Sprague-Dawley , Shab Potassium Channels/genetics , Signal Transduction/genetics
7.
J Neurochem ; 106(3): 1125-37, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18466331

ABSTRACT

Previously, we reported that apoptosis of cerebellar granular neurons induced by low-K+ and serum-free (LK-S) was associated with an increase in the A-type K+ channel current (I(A)), and an elevated expression of main alpha-subunit of the I(A) channel, which is known as Kv4.2 and Kv4.3. Here, we show, as assessed by quantitative RT-PCR and whole-cell recording, that besides Kv4.2 and Kv4.3, Kv1.1 is very important for I(A) channel. The expression of Kv1.1 was elevated in the apoptotic neurons, while silencing Kv1.1 expression by siRNA reduced the I(A) amplitude of the apoptotic neuron, and increased neuron viability. Inhibiting Kv1.1 current by dendrotoxin-K evoked a similar effect of reduction of I(A) amplitude and protection of neurons. Applying a protein kinase C (PKC) activator, phorbol ester acetate A (PMA) mimicked the LK-S-induced neuronal apoptotic effect, enhanced the I(A) amplitude and reduced the granule cell viability. The PKC inhibitor, bisindolylmaleimide I and Gö6976 protected the cell against apoptosis induced by LK-S. After silencing the Kv1.1 gene, the effect of PMA on the residual K+ current was reduced significantly. Quantitative RT-PCR and Western immunoblot techniques revealed that LK-S treatment and PMA increased the level of the expression of Kv1.1, in contrast, bisindolylmaleimide I inhibited Kv1.1 expression. In addition, the activation of the PKC isoform was identified in apoptotic neurons. We thus conclude that in the rat cerebellar granule cell, the I(A) channel associated with apoptotic neurons is encoded mainly by the Kv1.1 gene, and that the PKC pathway promotes neuronal apoptosis by a brief modulation of the I(A) amplitude and a permanent increase in the levels of expression of the Kv1.1 alpha-subunit.


Subject(s)
Apoptosis/physiology , Cerebellum/physiology , Kv1.1 Potassium Channel/biosynthesis , Neurons/physiology , Protein Kinase C/physiology , Animals , Animals, Newborn , Cell Survival/physiology , Cells, Cultured , Cerebellum/cytology , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/physiology , Neurons/cytology , Rats , Rats, Sprague-Dawley
8.
Neuropharmacology ; 51(4): 737-46, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16806301

ABSTRACT

Compelling evidence indicates that excessive potassium (K+) efflux and intracellular K+ depletion are the key early steps in apoptosis. Previously, we reported that apoptosis of cerebellar granule neurons induced by incubation in low-K+ (5 mM) and serum-free medium was associated with an increase in A-type transient inactivation of K+ channel current (IA) amplitude and modulation of channels' gating properties. Here, we showed that a classic K+ channel blocker, 4-aminopyradine (4-AP), significantly inhibited IA amplitude in a concentration-dependent manner (reduction of current by 10 microM and 10 mM 4-AP was 11.4+/-1.3% and 72.2+/-3.3%, respectively). Moreover, 4-AP modified the steady-state activation and inactivation kinetics of IA channels, such that the activation and inactivation curves were shifted to the right about 20 mV and 17 mV, respectively. Fluorescence staining showed that 4-AP dramatically increased the viability of cells undergoing apoptosis in a dose-dependent manner. That is, while 5 mM 4-AP was present, cell viability was 84.9+/-5.2%. Consistent with the cell viability analysis, internucleosomal DNA fragmentation by gel electrophoresis analysis showed that 5 mM 4-AP also protected against neuronal apoptosis. Furthermore, 4-AP significantly inhibited cytochrome c release and caspase-3 activity induced by low-K+/serum-free incubation. Finally, current-clamp analysis indicated that 5 mM 4-AP did not significantly depolarize the membrane potential. These results suggest that 4-AP has robust neuroprotective effects on apoptotic granule cells. The neuroprotective effect of 4-AP is likely not due to membrane depolarization, but rather that 4-AP may modulate the gating properties of IA channels in an anti-apoptotic manner.


Subject(s)
4-Aminopyridine/pharmacology , Apoptosis/drug effects , Cerebellum/cytology , Neurons/drug effects , Potassium Channel Blockers/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Patch-Clamp Techniques/methods , Potassium/pharmacology , Rats , Rats, Sprague-Dawley
9.
J Pineal Res ; 38(1): 53-61, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15617537

ABSTRACT

Compelling evidence indicates that excessive K+ efflux and intracellular K+ depletion are key early steps in apoptosis. Previously, we reported that apoptosis of cerebellar granular neurons induced by incubation under low K+ (5 mM) conditions was associated with an increase in delayed rectifier outward K+ current (IK) amplitude and caspase-3 activity. Moreover, the melatonin receptor antagonist 4P-PDOT abrogated the effects of 2-iodomelatonin on IK augmentation, caspase-3 activity and apoptosis. Here, we show that incubation under low K+/serum-free conditions for 6 hr led to a dramatic increase in the A-type transient outward K+ current (IA) (a 27% increase; n=31); in addition, fluorescence staining showed that under these conditions, cell viability decreased by 30% compared with the control. Treatment with 2-iodomelatonin inhibited the IA amplitude recorded from control and apoptotic cells in a concentration-dependent manner and modified the IA channel activation kinetics of cells under control conditions. Moreover, 2-iodomelatonin increased the viability of cell undergoing apoptosis. Interestingly, 4P-PDOT did not abrogate the effect of 2-iodomelatonin on IA augmentation under these conditions; in the presence of 4P-PDOT (100 microm), 2-iodomelatonin reduced the average IA by 41+/-4%, which was similar to the effect of 2-iodomelatonin alone. These results suggest that the neuroprotective effects of 2-idomelatonin are not only because of its antioxidant or receptor-activating properties, but rather that 2-iodomelatonin may inhibit IA channels by acting as a channel blocker.


Subject(s)
Apoptosis/drug effects , Cerebellum/drug effects , Cytoplasmic Granules/drug effects , Melatonin/analogs & derivatives , Melatonin/pharmacology , Neurons/drug effects , Animals , Cerebellum/cytology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
10.
Cell Motil Cytoskeleton ; 58(2): 127-36, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15083534

ABSTRACT

Cell migration is mediated by ion channels and transporters, and plays crucial roles in a variety of physiological and pathological processes. Previously, our studies have shown that a Ca(2+)-regulated K(+) current exists in B-16 murine melanoma cells, and that endothelin-1 (ET-1) inhibits the K(+) current via a PKC-dependent pathway. In the present study, patch-clamp whole-cell recording and transwell migration assays were used to examine the effects of ET-1 on B-16 murine melanoma cell migration. ET-1 (100 nM in the injection pipette and 10 nM in the incubation medium) decreased the K(+) current amplitude by 33.0 +/- 2.5% and inhibited migration of B-16 cells by 57.4 +/- 9.4%. Similarly, the Ca(2+)-regulated K(+) channel blockers, BaCl(2) and quinidine, decreased the K(+) current by 20.5 +/- 1.0% and 36.6 +/- 1.2%, respectively, and slowed migration of B-16 melanoma cells by 37.1 +/- 8.6% and 42.7 +/- 8.8%, respectively. The effect of ET-1 on the K(+) current and cell migration was simulated by ET-3. In contrast, the K(+) channel opener, diclofenac, increased the K(+) current by 128.8 +/- 11.7%, 257.4 +/- 35.8% at concentrations of 1 and 5 mM, respectively. Likewise, the migration of B-16 murine melanoma cells dramatically increased by 75.6 +/- 12.7% in the presence of 100 microM diclofenac in incubation medium. Furthermore, the ET-1- and ET-3-induced inhibition of K(+) current and migration was abrogated by diclofenac. In the presence of diclofenac, ET-1 only reduced the K(+) current amplitude by 10.6 +/- 1.1%, and slowed B-16 cell migration by only 10.8 +/- 8.9%. The results suggest that the K(+) channel-dependent migration of B-16 melanoma cells is modulated by ET-1. Cell Motil.


Subject(s)
Cell Movement/physiology , Endothelin-1/metabolism , Melanoma, Experimental/metabolism , Potassium/metabolism , Animals , Cell Movement/drug effects , Diclofenac/pharmacology , Endothelin-1/pharmacology , Melanoma, Experimental/pathology , Mice , Potassium/antagonists & inhibitors , Potassium Channels/drug effects
11.
J Pineal Res ; 36(2): 109-16, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14962062

ABSTRACT

Activation of K(+) current plays a critical role in the control of programmed cell death. In the present study, whole-cell patch-clamp recording, a caspase-3 activity assay, and flow cytometric analysis were used to examine the effects of the MT2 melatonin receptor agonist 2-iodomelatonin on the delayed-rectifier K(+) current (IK) and the prevention of apoptosis. It was found that apoptosis of cerebellar granular neurons induced by low-K(+) (5 mm) incubation was associated with an increase in IK amplitude and caspase-3 activity. After 6 hr of low-K(+) treatment, IK was increased by 45% (n = 86). Flow cytometry showed that the apoptosis rate increased by 333% compared with the control neurons. In addition, exposure of cultured granule cells to low K(+) also resulted in a significant activation of caspase-3, by 466%. 2-Iodomelatonin (10 microm in injection pipette) inhibited the IK amplitude recorded from control cells and from cells undergoing apoptosis. However, 2-iodomelatonin only modified the IK-channel activation kinetics of cells under both conditions. Furthermore, 2-iodomelatonin reduced the rate of apoptosis and caspase-3 activation, by 66 and 64%, respectively. The melatonin receptor antagonist, 4P-PDOT, abrogated the effect of 2-iodomelatonin on the IK augmentation, caspase-3 activity, and apoptosis. These results suggest that the neuroprotective effects of melatonin are not only because of its function as a powerful antioxidant, but also to its interactions with specific receptors. The effect of 2-iodomelatonin against apoptosis may be mediated by activating a melatonin receptor, which modulates IK channels and reduces K(+) efflux.


Subject(s)
Apoptosis/drug effects , Melatonin/analogs & derivatives , Melatonin/pharmacology , Neurons/drug effects , Receptors, Melatonin/agonists , Animals , Cerebellum/drug effects , Cerebellum/metabolism , Neurons/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...