Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Korean J Physiol Pharmacol ; 23(2): 121-130, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30820156

ABSTRACT

Glutamate toxicity-mediated mitochondrial dysfunction and neuronal cell death are involved in the pathogenesis of several neurodegenerative diseases as well as acute brain ischemia/stroke. In this study, we investigated the neuroprotective mechanism of dieckol (DEK), one of the phlorotannins isolated from the marine brown alga Ecklonia cava, against glutamate toxicity. Primary cortical neurons (100 µM, 24 h) and HT22 neurons (5 mM, 12 h) were stimulated with glutamate to induce glutamate toxic condition. The results demonstrated that DEK treatment significantly increased cell viability in a dose-dependent manner (1-50 µM) and recovered morphological deterioration in glutamate-stimulated neurons. In addition, DEK strongly attenuated intracellular reactive oxygen species (ROS) levels, mitochondrial overload of Ca2+ and ROS, mitochondrial membrane potential (ΔΨm) disruption, adenine triphosphate depletion. DEK showed free radical scavenging activity in the cell-free system. Furthermore, DEK enhanced protein expression of heme oxygenase-1 (HO-1), an important anti-oxidant enzyme, via the nuclear translocation of nuclear factor-like 2 (Nrf2). Taken together, we conclude that DEK exerts neuroprotective activities against glutamate toxicity through its direct free radical scavenging property and the Nrf-2/HO-1 pathway activation.

2.
Korean J Physiol Pharmacol ; 22(3): 311-319, 2018 May.
Article in English | MEDLINE | ID: mdl-29719453

ABSTRACT

Mitochondrial calcium overload is a crucial event in determining the fate of neuronal cell survival and death, implicated in pathogenesis of neurodegenerative diseases. One of the driving forces of calcium influx into mitochondria is mitochondria membrane potential (ΔΨm). Therefore, pharmacological manipulation of ΔΨm can be a promising strategy to prevent neuronal cell death against brain insults. Based on these issues, we investigated here whether nobiletin, a Citrus polymethoxylated flavone, prevents neurotoxic neuronal calcium overload and cell death via regulating basal ΔΨm against neuronal insult in primary cortical neurons and pure brain mitochondria isolated from rat cortices. Results demonstrated that nobiletin treatment significantly increased cell viability against glutamate toxicity (100 µM, 20 min) in primary cortical neurons. Real-time imaging-based fluorometry data reveal that nobiletin evokes partial mitochondrial depolarization in these neurons. Nobiletin markedly attenuated mitochondrial calcium overload and reactive oxygen species (ROS) generation in glutamate (100 µM)-stimulated cortical neurons and isolated pure mitochondria exposed to high concentration of Ca2+ (5 µM). Nobiletin-induced partial mitochondrial depolarization in intact neurons was confirmed in isolated brain mitochondria using a fluorescence microplate reader. Nobiletin effects on basal ΔΨm were completely abolished in K+-free medium on pure isolated mitochondria. Taken together, results demonstrate that K+ influx into mitochondria is critically involved in partial mitochondrial depolarization-related neuroprotective effect of nobiletin. Nobiletin-induced mitochondrial K+ influx is probably mediated, at least in part, by activation of mitochondrial K+ channels. However, further detailed studies should be conducted to determine exact molecular targets of nobiletin in mitochondria.

3.
J Neurosci Res ; 95(12): 2469-2482, 2017 12.
Article in English | MEDLINE | ID: mdl-28481020

ABSTRACT

A-type K+ channels (IA channels) contribute to learning and memory mechanisms by regulating neuronal excitabilities in the CNS, and their expression level is targeted by Ca2+ influx via synaptic NMDA receptors (NMDARs) during long-term potentiation (LTP). However, it is not clear how local synaptic Ca2+ changes induce IA downregulation throughout the neuron, extending from the active synapse to the soma. In this study, we tested if two major receptors of endoplasmic reticulum (ER), ryanodine (RyRs), and IP3 (IP3 R) receptors, are involved in Ca2+ -mediated IA downregulation in cultured hippocampal neurons of rats. The downregulation of IA channels was induced by doubling the Ca2+ concentration in culture media (3.6 mM for 24 hrs) or treating with glycine (200 µM for 3 min) to induce chemical LTP (cLTP), and the changes in IA peaks were measured electrophysiologically by a whole-cell patch. We confirmed that Ca2+ or glycine treatment significantly reduced IA peaks and that their effects were abolished by blocking NMDARs or voltage-dependent Ca2+ channels (VDCCs). In this cellular processing, blocking RyRs (by ryanodine, 10 µM) but not IP3 Rs (by 2APB, 100 µM) completely abolished IA downregulation, and the LTP observed in hippocampal slices was more diminished by ryanodine rather than 2APB. Furthermore, blocking RyRs also reduced Ca2+ -mediated PKA activation, indicating that sequential signaling cascades, including the ER and PKA, are involved in regulating IA downregulation. These results strongly suggest a possibility that RyR contribution and mediated IA downregulation are required to regulate membrane excitability as well as synaptic plasticity in CA3-CA1 connections of the hippocampus. © 2017 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley
4.
Korean J Physiol Pharmacol ; 21(2): 259-265, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28280420

ABSTRACT

Excessive influx and the subsequent rapid cytosolic elevation of Ca2+ in neurons is the major cause to induce hyperexcitability and irreversible cell damage although it is an essential ion for cellular signalings. Therefore, most neurons exhibit several cellular mechanisms to homeostatically regulate cytosolic Ca2+ level in normal as well as pathological conditions. Delayed rectifier K+ channels (IDR channels) play a role to suppress membrane excitability by inducing K+ outflow in various conditions, indicating their potential role in preventing pathogenic conditions and cell damage under Ca2+-mediated excitotoxic conditions. In the present study, we electrophysiologically evaluated the response of IDR channels to hyperexcitable conditions induced by high Ca2+ pretreatment (3.6 mM, for 24 hours) in cultured hippocampal neurons. In results, high Ca2+-treatment significantly increased the amplitude of IDR without changes of gating kinetics. Nimodipine but not APV blocked Ca2+-induced IDR enhancement, confirming that the change of IDR might be targeted by Ca2+ influx through voltage-dependent Ca2+ channels (VDCCs) rather than NMDA receptors (NMDARs). The VDCC-mediated IDR enhancement was not affected by either Ca2+-induced Ca2+ release (CICR) or small conductance Ca2+-activated K+ channels (SK channels). Furthermore, PP2 but not H89 completely abolished IDR enhancement under high Ca2+ condition, indicating that the activation of Src family tyrosine kinases (SFKs) is required for Ca2+-mediated IDR enhancement. Thus, SFKs may be sensitive to excessive Ca2+ influx through VDCCs and enhance IDR to activate a neuroprotective mechanism against Ca2+-mediated hyperexcitability in neurons.

5.
J Nanosci Nanotechnol ; 16(2): 2065-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27433729

ABSTRACT

We have developed the first nanoengineered quantum dot molecular complex designed to measure changes of calcium ion (Ca2+) concentration at high spatial and temporal resolutions in real time. The sensor is ratiometric and composed of three components: a quantum dot (QD) emitting at 620 nm as a fluorescence donor, an organic dye (Alexa Fluor 647) as a fluorescence acceptor, and a calmodulin-M13 (CaM-M13) protein part as a calcium sensing component. In this work, we have determined the maximal number of CaM-M13 required for saturating a single QD particle to be approximately 16. The dissociation constant, Kd of the QD-based calcium ion sensor was also estimated to be around 30 microM.

6.
Korean J Physiol Pharmacol ; 19(3): 219-28, 2015 May.
Article in English | MEDLINE | ID: mdl-25954126

ABSTRACT

Excessive microglial activation and subsequent neuroinflammation lead to synaptic loss and dysfunction as well as neuronal cell death, which are involved in the pathogenesis and progression of several neurodegenerative diseases. Thus, the regulation of microglial activation has been evaluated as effective therapeutic strategies. Although dieckol (DEK), one of the phlorotannins isolated from marine brown alga Ecklonia cava, has been previously reported to inhibit microglial activation, the molecular mechanism is still unclear. Therefore, we investigated here molecular mechanism of DEK via extracellular signal-regulated kinase (ERK), Akt and nicotinamide adenine dinuclelotide phosphate (NADPH) oxidase-mediated pathways. In addition, the neuroprotective mechanism of DEK was investigated in microglia-mediated neurotoxicity models such as neuron-microglia co-culture and microglial conditioned media system. Our results demonstrated that treatment of anti-oxidant DEK potently suppressed phosphorylation of ERK in lipopolysaccharide (LPS, 1 µg/ml)-stimulated BV-2 microglia. In addition, DEK markedly attenuated Akt phosphorylation and increased expression of gp91 (phox) , which is the catalytic component of NADPH oxidase complex responsible for microglial reactive oxygen species (ROS) generation. Finally, DEK significantly attenuated neuronal cell death that is induced by treatment of microglial conditioned media containing neurotoxic secretary molecules. These neuroprotective effects of DEK were also confirmed in a neuron-microglia co-culture system using enhanced green fluorescent protein (EGFP)-transfected B35 neuroblastoma cell line. Taken together, these results suggest that DEK suppresses excessive microglial activation and microglia-mediated neuronal cell death via downregulation of ERK, Akt and NADPH oxidase-mediated pathways.

7.
Complement Ther Med ; 22(3): 456-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24906585

ABSTRACT

OBJECTIVES: Aromatherapy massage is commonly used for the stress management of healthy individuals, and also has been often employed as a therapeutic use for pain control and alleviating psychological distress, such as anxiety and depression, in oncological palliative care patients. However, the exact biological basis of aromatherapy massage is poorly understood. Therefore, we evaluated here the effects of aromatherapy massage interventions on multiple neurobiological indices such as quantitative psychological assessments, electroencephalogram (EEG) power spectrum pattern, salivary cortisol and plasma brain-derived neurotrophic factor (BDNF) levels. DESIGN: A control group without treatment (n = 12) and aromatherapy massage group (n = 13) were randomly recruited. They were all females whose children were diagnosed as attention deficit hyperactivity disorder and followed up in the Department of Psychiatry, Jeju National University Hospital. Participants were treated with aromatherapy massage for 40 min twice per week for 4 weeks (8 interventions). RESULTS: A 4-week-aromatherapy massage program significantly improved all psychological assessment scores in the Stat-Trait Anxiety Index, Beck Depression Inventory and Short Form of Psychosocial Well-being Index. Interestingly, plasma BDNF levels were significantly increased after a 4 week-aromatherapy massage program. Alpha-brain wave activities were significantly enhanced and delta wave activities were markedly reduced following the one-time aromatherapy massage treatment, as shown in the meditation and neurofeedback training. In addition, salivary cortisol levels were significantly reduced following the one-time aromatherapy massage treatment. CONCLUSIONS: These results suggest that aromatherapy massage could exert significant influences on multiple neurobiological indices such as EEG pattern, salivary cortisol and plasma BDNF levels as well as psychological assessments.


Subject(s)
Aromatherapy , Brain Waves/physiology , Brain-Derived Neurotrophic Factor/blood , Hydrocortisone/analysis , Massage , Stress, Psychological/therapy , Adult , Electroencephalography , Female , Humans , Middle Aged , Saliva/chemistry
8.
Korean J Physiol Pharmacol ; 18(2): 135-41, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24757375

ABSTRACT

The downregulation of A-type K(+) channels (IA channels) accompanying enhanced somatic excitability can mediate epileptogenic conditions in mammalian central nervous system. As IA channels are dominantly targeted by dendritic and postsynaptic processings during synaptic plasticity, it is presumable that they may act as cellular linkers between synaptic responses and somatic processings under various excitable conditions. In the present study, we electrophysiologically tested if the downregulation of somatic IA channels was sensitive to synaptic activities in young hippocampal neurons. In primarily cultured hippocampal neurons (DIV 6~9), the peak of IA recorded by a whole-cell patch was significantly reduced by high KCl or exogenous glutamate treatment to enhance synaptic activities. However, the pretreatment of MK801 to block synaptic NMDA receptors abolished the glutamate-induced reduction of the IA peak, indicating the necessity of synaptic activation for the reduction of somatic IA. This was again confirmed by glycine treatment, showing a significant reduction of the somatic IA peak. Additionally, the gating property of IA channels was also sensitive to the activation of synaptic NMDA receptors, showing the hyperpolarizing shift in inactivation kinetics. These results suggest that synaptic LTP possibly potentiates somatic excitability via downregulating IA channels in expression and gating kinetics. The consequential changes of somatic excitability following the activity-dependent modulation of synaptic responses may be a series of processings for neuronal functions to determine outputs in memory mechanisms or pathogenic conditions.

9.
Neurosci Bull ; 30(3): 505-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24526657

ABSTRACT

In the mammalian brain, information encoding and storage have been explained by revealing the cellular and molecular mechanisms of synaptic plasticity at various levels in the central nervous system, including the hippocampus and the cerebral cortices. The modulatory mechanisms of synaptic excitability that are correlated with neuronal tasks are fundamental factors for synaptic plasticity, and they are dependent on intracellular Ca(2+)-mediated signaling. In the present review, the A-type K(+) (IA) channel, one of the voltage-dependent cation channels, is considered as a key player in the modulation of Ca(2+) influx through synaptic NMDA receptors and their correlated signaling pathways. The cellular functions of IA channels indicate that they possibly play as integral parts of synaptic and somatic complexes, completing the initiation and stabilization of memory.


Subject(s)
Hippocampus/cytology , Kv Channel-Interacting Proteins/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/metabolism , Animals , Neurons/cytology
10.
Korean J Physiol Pharmacol ; 18(6): 457-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25598658

ABSTRACT

At central synapses, activity-dependent synaptic plasticity has a crucial role in information processing, storage, learning, and memory under both physiological and pathological conditions. One widely accepted model of learning mechanism and information processing in the brain is Hebbian Plasticity: long-term potentiation (LTP) and long-term depression (LTD). LTP and LTD are respectively activity-dependent enhancement and reduction in the efficacy of the synapses, which are rapid and synapse-specific processes. A number of recent studies have a strong focal point on the critical importance of another distinct form of synaptic plasticity, non-Hebbian plasticity. Non-Hebbian plasticity dynamically adjusts synaptic strength to maintain stability. This process may be very slow and occur cell-widely. By putting them all together, this mini review defines an important conceptual difference between Hebbian and non-Hebbian plasticity.

11.
Phytother Res ; 27(4): 564-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22678994

ABSTRACT

Mitochondrial membrane potential (∆Ψm ) contributes to determining a driving force for calcium to enter the mitochondria. It has been demonstrated that even a small mitochondrial depolarization is sufficient to prevent mitochondrial calcium overload and the subsequent apoptosis. Therefore, mild mitochondrial depolarization has been recently evaluated as a novel mechanism of neuroprotection via inhibiting neurotoxic mitochondrial calcium overload during neuronal insults. In the present study, using both real-time recording and flow cytometric analyses of ∆Ψm , we demonstrated that ethanolic peel extract of Citrus sunki Hort. ex Tanaka (CPE) and its active compounds are capable of inducing a mild mitochondrial depolarization. Polymethoxylated flavones such as nobiletin and tangeretin were found as the active compounds responsible for CPE effects on ∆Ψm . Neuronal viability was significantly increased in a dose-dependent manner by CPE treatment in H2 O2 -stimulated HT-22 cells as an in vitro neuronal insult model. CPE treatment significantly inhibited H2 O2 -induced apoptotic processes such as chromatin condensation, caspase 3 activation and anti-poly (ADP-ribose) polymerase (PARP) cleavage. CPE treatment significantly blocked mitochondrial calcium overload in H2 O2 -stimulated HT-22 neurons as indicated by rhod-2 acetoxymethyl ester. Taken together, our findings suggest that CPE and its active compounds may be considered as promising neuroprotective agents via inducing a mild mitochondrial depolarization.


Subject(s)
Citrus/chemistry , Membrane Potential, Mitochondrial/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Apoptosis , Calcium/metabolism , Cell Line , Flavones/pharmacology , Flow Cytometry , Fruit/chemistry , Humans , Hydrogen Peroxide/pharmacology , Mitochondria/drug effects
12.
Brain Res Bull ; 91: 14-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219935

ABSTRACT

Immature hippocampal neurons with high input resistances (R(in)) are vulnerable to hyperexcitable or epileptogenic conditions. This phenomenon has been suggested to explain the neuroprotective roles of hyperpolarization-activated cation channels (I(h) channels) to regulate membrane R(in). In the present study, we tried to electrophysiologically clarify the relationship between membrane R(in) and I(h) channels and determine the neuroprotective roles of these channels in development. The CA1 neurons from rats (within 3 postnatal weeks) were classified into two groups based on the onset time (shorter or longer than 20 ms) to fire the first action potential (AP) in response to a current injection (100 pA, 800 ms). Neurons with a shorter onset time (Short-OsT), exhibited higher R(in), while neurons with longer onset times (Long-OsT) revealed lower R(in). Unexpectedly, Short-OsT neurons with higher R(in) exhibited larger amplitudes of I(h) compared with Long-OsT neurons. Furthermore, the application of temporal depolarization stimulus (TDS, -14 mV holding for 150 s) significantly enhanced suprathreshold excitabilities of repetitive APs in Long-OsT but not Short-OsT neurons, suggesting a protective role of I(h) channels under high R(in) conditions. In the presence of the specific hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker ZD7288, TDS also enhanced the excitability of Short-OsT neurons, suggesting that young CA1 neurons regulate I(h) channel expression for neuroprotective modulation against epileptogenic conditions.


Subject(s)
Ion Channels/metabolism , Pyramidal Cells/metabolism , Action Potentials/physiology , Animals , CA1 Region, Hippocampal/metabolism , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
13.
Korean J Physiol Pharmacol ; 16(5): 343-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23118559

ABSTRACT

Blocking or regulating K(+) channels is important for investigating neuronal functions in mammalian brains, because voltage-dependent K(+) channels (Kv channels) play roles to regulate membrane excitabilities for synaptic and somatic processings in neurons. Although a number of toxins and chemicals are useful to change gating properties of Kv channels, specific effects of each toxin on a particular Kv subunit have not been sufficiently demonstrated in neurons yet. In this study, we tested electrophysiologically if heteropodatoxin2 (HpTX(2)), known as one of Kv4-specific toxins, might be effective on various K(+) outward currents in CA1 neurons of organotypic hippocampal slices of rats. Using a nucleated-patch technique and a pre-pulse protocol in voltage-clamp mode, total K(+) outward currents recorded in the soma of CA1 neurons were separated into two components, transient and sustained currents. The extracellular application of HpTX(2) weakly but significantly reduced transient currents. However, when HpTX(2) was added to internal solution, the significant reduction of amplitudes were observed in sustained currents but not in transient currents. This indicates the non-specificity of HpTX(2) effects on Kv4 family. Compared with the effect of cytosolic 4-AP to block transient currents, it is possible that cytosolic HpTX(2) is pharmacologically specific to sustained currents in CA1 neurons. These results suggest that distinctive actions of HpTX(2) inside and outside of neurons are very efficient to selectively reduce specific K(+) outward currents.

14.
Eur J Pharmacol ; 690(1-3): 4-12, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22683871

ABSTRACT

Excessive microglial activation-mediated neurotoxicity has been implicated in playing a crucial role in the pathogenesis of stroke and neurodegenerative diseases. Therefore, much attention has been paid to therapeutic strategies aimed at suppressing neurotoxic microglial activation. The microglial regulatory mechanism of methyl lucidone, a cyclopentenedione isolated from the stem bark of Lindera erythrocarpa Makino, was investigated in the present study. Methyl lucidone treatment (0.1-10 µM) significantly inhibited lipopolysaccharide (LPS, 100 ng/ml, 24 h)-stimulated nitric oxide (NO) production in a dose-dependent manner in both primary cortical microglia and BV-2 cell line. Moreover, it strongly inhibited LPS-stimulated secretion of pro-inflammatory cytokines, such as interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α). Methyl lucidone treatment markedly induced down-regulation of LPS-induced nuclear translocation of nuclear factor κB (NF-κB) through preventing the degradation of the inhibitory protein IκBα. In addition, phosphorylation of Akt and mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK) and p38 kinases were also suppressed by methyl lucidone. The cell viabilities of HT-22 neurons were significantly attenuated by treatment of the conditioned media containing neurotoxic secretary molecules from LPS-stimulated microglia. However, methyl lucidone significantly blocked neuronal cell death induced by microglial conditioned media. These neuroprotective effects of methyl lucidone were also confirmed in a neuron-microglia co-culture system using EGFP-transfected B35 neuroblastoma cell line. Taken together, these results suggest that methyl lucidone may have a neuroprotective potential via inhibition of neurotoxic microglial activation implicated in neurodegeneration.


Subject(s)
Cyclopentanes/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Death/drug effects , Cerebral Cortex/cytology , Coculture Techniques , Culture Media, Conditioned/metabolism , Cytokines/metabolism , Lindera/chemistry , Lipopolysaccharides/pharmacology , Microglia/cytology , Microglia/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nitric Oxide/biosynthesis , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects
15.
Brain Res Bull ; 84(1): 17-21, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20920553

ABSTRACT

Proper expression of synaptic NMDA receptors (NMDARs) is necessary to regulate synaptic Ca²(+) influx and the induction the long-term potentiation (LTP) in the mammalian hippocampus. Previously we reported that expressing the A-type K(+) channel subunit Kv4.2 in CA1 neurons of organotypic slice cultures reduced synaptic NR2B-containing NMDAR expression and completely blocked LTP induced by a pairing protocol. As pretreatment with an NMDAR antagonist (APV) overnight blocked the reduction of NR2B-containing receptors in neurons expressing EGFP-labeled Kv4.2 (Kv4.2g), we hypothesized that LTP would be rescued in Kv4.2g neurons by overnight treatment with APV. We report here that the overnight APV pretreatment in Kv4.2g-expressing neurons only partially restored potentiation. This partial potentiation was completely blocked by inhibition of the CAMKII kinase. These results indicate that A-type K(+) channels must regulate synaptic integration and plasticity through another mechanism in addition to their regulation of synaptic NR2 subunit composition. We suggest that dendritic excitability, which is regulated by Kv4.2 expression, also contributes to synaptic plasticity.


Subject(s)
Long-Term Potentiation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Shal Potassium Channels/metabolism , Synapses/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/cytology , Hippocampus/physiology , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Potassium Channel Blockers/metabolism , Rats , Rats, Sprague-Dawley , Shal Potassium Channels/genetics , Tissue Culture Techniques , Valine/analogs & derivatives , Valine/metabolism
16.
Neurochem Res ; 36(2): 223-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21057871

ABSTRACT

Recently, it was reported that in a 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model, neuronal cell death is associated with the cdk5-mediated hyperphosphorylation of myocyte enhancer factor 2 (MEF2), a transcription factor that is critically required for neuronal survival. In the present study, we investigated the possible involvement of cdk5-mediated MEF2D down-regulation on 6-hydroxydopamine (6-OHDA)-induced PC12 cell death. 6-OHDA was found to significantly increase nitric oxide (NO) production and to induce apoptosis in a time-dependent manner in PC12 cells. Furthermore, 6-OHDA was found to markedly reduce MEF2D levels under conditions that could induce PC12 cell apoptosis. In addition, PC12 cell death and MEF2D degradation by 6-OHDA were prevented by the cdk5 inhibitor roscovitine, but roscovitine could not restore the 6-OHDA-induced inactivation of Akt. These results suggest that the cell death and MEF2D degradation caused by 6-OHDA are dependent on cdk5 activity. On the other hand, roscovitine enhanced the 6-OHDA-induced activations of ERK1/2 and JNK, but reduced the 6-OHDA-induced activation of p38. These results suggest that PC12 cell death by 6-OHDA appears to be regulated by the down-regulation of MEF2D via some interaction between cdk5 and MAP kinase.


Subject(s)
Apoptosis/drug effects , Myogenic Regulatory Factors/metabolism , Oxidopamine/pharmacology , PC12 Cells/drug effects , PC12 Cells/physiology , Animals , Apoptosis/physiology , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , DNA Fragmentation , Down-Regulation , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , MEF2 Transcription Factors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myogenic Regulatory Factors/genetics , Nitric Oxide/metabolism , Protein Kinase Inhibitors/metabolism , Purines/metabolism , Rats , Roscovitine , Signal Transduction/physiology
17.
Biol Pharm Bull ; 33(11): 1814-21, 2010.
Article in English | MEDLINE | ID: mdl-21048305

ABSTRACT

A growing body of evidence suggests that nobiletin (5,6,7,8,3',4'-hexamethoxy flavone) from the peel of citrus fruits, enhances the damaged cognitive function in disease animal models. However, the neuroprotective mechanism has not been clearly elucidated. Since nobiletin has shown anti-inflammatory effects in several tissues, we investigated whether nobiletin suppresses excessive microglial activation implicated in neurotoxicity in lipopolysaccharide (LPS)-stimulated BV-2 microglia cell culture models. Release of nitric oxide (NO), the major inflammatory mediator in microglia, was markedly suppressed in a dose-dependent manner following nobiletin treatment (1-50 µM) in LPS-stimulated BV-2 microglia cells. The inhibitory effect of nobiletin was similar to that of minocycline, a well-known microglial inactivator. Nobiletin significantly inhibited the release of the pro-inflammatory cytokine tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß). LPS-induced phosphorylations of extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal kinase (JNK), and p38 mitogen-activated protein kinases (MAPKs) were also significantly inhibited by nobiletin treatment. In addition, nobiletin markedly inhibited the LPS-induced pro-inflammatory transcription factor nuclear factor κB (NF-κB) signaling pathway by suppressing nuclear NF-κB translocation from the cytoplasm and subsequent expression of NF-κB in the nucleus. Taken together, these results may contribute to further exploration of the therapeutic potential and molecular mechanism of nobiletin in relation to neuroinflammation and neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Citrus/chemistry , Inflammation/drug therapy , Microglia/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytokines/antagonists & inhibitors , Cytoplasm/drug effects , Dose-Response Relationship, Drug , Fruit , Inflammation/metabolism , Lipopolysaccharides , Mice , Microglia/metabolism , Minocycline/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Nitric Oxide/antagonists & inhibitors , Phosphorylation , Phytotherapy , Signal Transduction/drug effects
18.
Korean J Urol ; 51(2): 94-100, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20414420

ABSTRACT

PURPOSE: The fibroblast growth factor receptor 3 (FGFR3) gene is known to be frequently mutated in noninvasive urothelial carcinomas of the bladder. In this study, we investigated the expression of FGFR3, Ki-67, and p53 in bladder cancers and the effects of expression on tumor recurrence. MATERIALS AND METHODS: Fifty-five cases of primary bladder cancer were examined by immunohistochemistry. The relationship of these markers with various clinicopathological factors, including recurrence, was assessed. RESULTS: Positivity for cytoplasmic FGFR3 (FGFR3-c) was associated with a lower cancer grade (p=0.022) and stage (p=0.011). Recurrence was more frequent in patients with a higher stage, negative FGFR3-c, and high Ki-67 expression. According to univariate analysis, predictors of recurrence-free survival included the following: age, stage, FGFR-c, Ki-67, and p53. However, none of these was independent from the other parameters in multivariate studies. CONCLUSIONS: The immunohistochemical expression of FGFR3 is not only one of the characteristic features of lower-grade and lower-stage urothelial carcinoma but also a possible marker in predicting disease recurrence.

19.
J Toxicol Environ Health A ; 71(20): 1392-9, 2008.
Article in English | MEDLINE | ID: mdl-18704831

ABSTRACT

The objective of the present study was to determine the mechanism by which KIOM-79 induced heme oxygenase-1 (HO-1) in rat pancreatic beta-cells (RINm5F). A mixture of plant extracts (KIOM-79) was obtained from Magnolia officinalis, Pueraria lobata, Glycyrrhiza uralensis, and Euphorbia pekinensis. HO-1, an antioxidant phase 2 enzyme, was previously reported to possess cytoprotective properties in pancreatic beta-cells. KIOM-79 induced heme oxygenase-1 (HO-1) expression at the mRNA and protein levels, leading to increased HO-1 activity. The transcription factor, NF-E2 related factor 2 (Nrf2), regulates the antioxidant response element (ARE) of the phase 2 detoxifying and antioxidant enzymes, resulting in modulation of HO-1 expression. KIOM-79 increased nuclear translocation, ARE binding, and transcriptional activity of Nrf2. Furthermore, KIOM-79 also elicited activation of Akt (protein kinase B) and LY294004 (inhibitor of Akt)-suppressed KIOM-79-induced activation of Nrf2, which subsequently decreased HO-1 protein levels. Taken together, these data suggest that KIOM-79 augments the cellular antioxidant defense capacity through induction of HO-1 via the Akt-Nrf2-ARE signaling pathway, thereby protecting cells from streptozotocin-induced oxidative stress.


Subject(s)
Heme Oxygenase (Decyclizing)/biosynthesis , Insulin-Secreting Cells/drug effects , NF-E2-Related Factor 2/drug effects , Plant Extracts/pharmacology , Streptozocin/antagonists & inhibitors , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Insulin-Secreting Cells/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/physiology , Oxidative Stress/drug effects , Rats , Reverse Transcriptase Polymerase Chain Reaction , Streptozocin/toxicity
20.
Cell Biol Int ; 32(9): 1099-107, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18550394

ABSTRACT

We have elucidated the cytoprotective effect of annphenone (2,4-dihyroxy-6-methoxy-acetophenone 4-O-beta-D-glucopyranoside) against oxidative stress-induced apoptosis. Annphenone scavenged intracellular reactive oxygen species (ROS) and increased antioxidant enzyme activities. It thereby prevented lipid peroxidation and DNA damage, which was demonstrated by the inhibition of the formation of thiobarbituric acid reactive substance (TBARS), inhibition of the comet tail and decreased phospho-H2A.X expression. Annphenone protected Chinese hamster lung fibroblast (V79-4) cells from cell death via the inhibition of apoptosis induced by hydrogen peroxide (H2O2), as shown by decreased apoptotic nuclear fragmentation, decreased sub-G1 cell population and inhibited mitochondrial membrane potential (Deltapsi) loss. Taken together, these findings suggest that annphenone exhibits antioxidant properties by inhibiting ROS generation and thus protecting cells from H2O2-induced cell damage.


Subject(s)
Acetophenones/pharmacology , Apoptosis/drug effects , Cytoprotection/drug effects , Fibroblasts/cytology , Glucosides/pharmacology , Hydrogen Peroxide/pharmacology , Lung/cytology , Oxidative Stress/drug effects , Acetophenones/chemistry , Animals , Antioxidants/pharmacology , Cell Line , Cricetinae , DNA Damage , Fibroblasts/drug effects , Fibroblasts/enzymology , Free Radical Scavengers/pharmacology , Glucosides/chemistry , Intracellular Space/drug effects , Intracellular Space/metabolism , Lipid Peroxidation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...