Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Neurosci Res ; 91(6): 745-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23553806

ABSTRACT

Spinal astrocytes have important mechanistic contributions to the initiation and maintenance of neurodegenerative diseases and chronic pain. Under inflammatory conditions, spinal astrocytes are exposed to cytokines such as tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and these cytokines could alter astrocytic function by modulating connexin (Cx43), subunits that form channels that modulate intercellular communication in astrocytes. The current study investigated the alteration of Cx43-gap junction in rat primary cultured spinal astrocytes stimulated with cytokines by real-time PCR and Western blotting. The transcriptional and translational levels of Cx43 were significantly but partially reduced 24 and 48 hr treatment with either TNF-α (10 ng/ml) or IFN-γ (5 ng/ml). A mixture of TNF-α and IFN-γ led to a robust decrease of Cx43 expression and, moreover, a moderate reduction of gap junction intercellular communication (GJIC), which was evaluated by a scrap loading/dye transfer assay. Both the decrease of Cx43 expression and the reduction in GJIC induced by the mixture of TNF-α and IFN-γ were prevented by blocking c-jun terminal kinase (JNK) but not by blocking extracellular signaling molecules ERK and p38 kinase, indicating a specific role of astrocytic JNK in the response to cytokines. In addition, treatment with cytokines potently induced the phosphorylation of JNK and c-jun in a time-dependent manner. These results indicate that intercellular communication of astrocytes is significantly disrupted in the inflammatory state and that stimulation of spinal astrocytes with inflammatory cytokines leads to significant inhibition of Cx43-GJIC through activation of the JNK signaling pathway.


Subject(s)
Astrocytes/metabolism , Cell Communication/physiology , Connexin 43/biosynthesis , Interferon-gamma/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Astrocytes/cytology , Blotting, Western , Cell Communication/drug effects , Cells, Cultured , Gap Junctions/metabolism , Inflammation/metabolism , Interferon-gamma/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Spinal Cord/cytology , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/pharmacology
2.
Neurochem Int ; 60(8): 817-26, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22446583

ABSTRACT

Spinal astrocytes have key roles in the regulation of pain transmission. However, the relationship between astrocytes and the circadian system in the spinal cord remains poorly defined. In the current study, the circadian variations in the expression of several clock genes in the lumbar spinal cord of mice were examined by using real-time PCR. The expression of Period1, Period2 and Cryptochrome1 showed significant circadian oscillations, each gene peaking in the early evening. The expression of Bmal1 mRNA also exhibited a circadian pattern, peaking from around midnight to early morning. The mRNA levels of Cryptochrome2 were slightly, but not significantly altered. Molecules related to pain transmission were also investigated. The mRNA expression of glutamine synthase (GS), and cyclooxygenases (COXs), known to be involved in various spinal sensory functions, showed rhythmicity with a peak in the early evening, although the expression of the neurokinin-1 receptor, subunits of the N-methyl-d-aspartate receptor, and glutamate transporters did not change. In addition, we found that protein levels of GS and COX-1 were also high at midnight compared with midday. Furthermore, we examined the effect of intrathecal fluorocitrate (100pmol), an inhibitor of astrocytic metabolism, on the expression of oscillating genes in lumbar spinal cord. Fluorocitrate significantly suppressed astrocyte function. Furthermore, the circadian oscillation of clock gene expression and GS and COX-1 expression were suppressed. Together, these results suggest that a significant circadian rhythmicity of the expression of clock genes is present in the spinal cord and that the components of the circadian clock timed by astrocytes might contribute to spinal functions, including nociceptive processes.


Subject(s)
Astrocytes/cytology , CLOCK Proteins/metabolism , Circadian Rhythm , Cyclooxygenase 1/metabolism , Glutamate-Ammonia Ligase/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Animals , Base Sequence , DNA Primers , Immunohistochemistry , Lumbar Vertebrae , Mice , Real-Time Polymerase Chain Reaction
3.
J Neurochem ; 120(6): 1036-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22182301

ABSTRACT

To examine mechanisms underlying substance P (SP) release from primary sensory neurons in response to activation of the non-selective cation channel transient receptor potential ankyrin 1 (TRPA1), SP release from cultured rat dorsal root ganglion neurons was measured, using radioimmunoassay, by stimulating TRPA1 with allyl isothiocyanate (AITC), a TRPA1 agonist. AITC-evoked SP release occurred in a concentration- and time-dependent manner. Interestingly, p38 mitogen-activated protein kinase (p38) inhibitor SB203580 significantly attenuated AITC-evoked SP release. The in vivo effect of AITC-evoked SP release from primary sensory neurons in mice was evaluated. Hind paw intraplantar injection of AITC induced nociceptive behaviors and inflammation (edema, thermal hyperalgesia). AITC-induced thermal hyperalgesia and edema were inhibited by intraplantar pre-treatment with either SB203580 or neurokinin-1 receptor antagonist CP96345. Moreover, intrathecal pre-treatment with either CP96345 or SB203580 inhibited AITC-induced nociceptive behaviors and thermal hyperalgesia. Immunohistochemical studies demonstrated that intraplantar AITC injection induced the phosphorylation of p38 in mouse dorsal root ganglion neurons containing SP. These findings suggest that activation of TRPA1 evokes SP release from the primary sensory neurons through phosphorylation of p38, subsequent nociceptive behaviors and inflammatory responses. Furthermore, the data also indicate that blocking the effects of TRPA1 activation at the periphery leads to significant antinociception.


Subject(s)
Receptors, Neurokinin-1/metabolism , Sensory Receptor Cells/metabolism , Substance P/metabolism , TRPC Cation Channels/metabolism , Acetanilides/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Inflammation/chemically induced , Isothiocyanates/adverse effects , Isothiocyanates/pharmacology , Male , Nociception/drug effects , Phosphorylation/drug effects , Protein Transport/drug effects , Purines/pharmacology , Radioimmunoassay , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , TRPA1 Cation Channel , TRPC Cation Channels/agonists , TRPC Cation Channels/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Brain Res ; 1410: 132-40, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21803334

ABSTRACT

OBJECTIVE: Although electroconvulsive therapy (ECT) is a well-established treatment for psychiatric disorders, its mechanism of action remains unclear. To investigate the cerebral hemodynamic response during ECT, we measured the changes in the regional cerebral blood flow (rCBF) at the bilateral prefrontal cortex (PFC) using near-infrared spectroscopy (NIRS). Method. The participants included eleven patients with schizophrenia and ten patients with mood disorders. The normalized tissue hemoglobin index (nTHI) was used as a sensitive parameter of rCBF by the SRS method and was measured during bilateral ECT using a two-channel NIRS. Results. 1. All patients responded to ECT treatment. 2. The levels of bilateral nTHI indicated a transient decrease during electrical stimulation and immediately were increased at both ictal and post-ictal phases by approximately 20% above baseline. 3. Patients with schizophrenia, but not mood disorders, showed significant asymmetric alteration of nTHI levels (left>right) during both the ictal and post-ictal phases. 4. The asymmetry index of nTHI, which indicates the difference between the left and right sides of the nTHI, was negatively correlated with the period of illness for schizophrenia, although the asymmetry index was not significantly correlated with any other clinical data, such as the effect of ECT treatment. Conclusion. Preliminary data demonstrated that bilateral ECT caused hemodynamic changes in bilateral PFC, and asymmetric alteration was found for schizophrenia, but not for mood disorders. Although further studies are necessary, the asymmetric hemodynamic response by ECT may be associated with the pathophysiology of schizophrenia, especially in the early stages.


Subject(s)
Cerebrovascular Circulation/physiology , Electroconvulsive Therapy , Hemodynamics/physiology , Prefrontal Cortex/physiopathology , Schizophrenia/physiopathology , Adult , Aged , Female , Humans , Male , Middle Aged , Mood Disorders/physiopathology , Mood Disorders/therapy , Prefrontal Cortex/blood supply , Schizophrenia/therapy , Spectroscopy, Near-Infrared
5.
J Biol Chem ; 286(24): 21118-28, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21515689

ABSTRACT

Recently, both clinical and animal studies demonstrated neuronal and glial plasticity to be important for the therapeutic action of antidepressants. Antidepressants increase glial cell line-derived neurotrophic factor (GDNF) production through monoamine-independent protein-tyrosine kinase, extracellular signal-regulated kinase (ERK), and cAMP responsive element-binding protein (CREB) activation in glial cells (Hisaoka, K., Takebayashi, M., Tsuchioka, M., Maeda, N., Nakata, Y., and Yamawaki, S. (2007) J. Pharmacol. Exp. Ther. 321, 148-157; Hisaoka, K., Maeda, N., Tsuchioka, M., and Takebayashi, M. (2008) Brain Res. 1196, 53-58). This study clarifies the type of tyrosine kinase and mechanism of antidepressant-induced GDNF production in C6 glioma cells and normal human astrocytes. The amitriptyline (a tricyclic antidepressant)-induced ERK activation was specifically and completely inhibited by fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitors and siRNA for FGFR1 and -2. Treatment with amitriptyline or several different classes of antidepressants, but not non-antidepressants, acutely increased the phosphorylation of FGFRs and FGFR substrate 2α (FRS2α). Amitriptyline-induced CREB phosphorylation and GDNF production were blocked by FGFR-tyrosine kinase inhibitors. Therefore, antidepressants activate the FGFR/FRS2α/ERK/CREB signaling cascade, thus resulting in GDNF production. Furthermore, we attempted to elucidate how antidepressants activate FGFR signaling. The effect of amitriptyline was inhibited by heparin, non-permeant FGF-2 neutralizing antibodies, and matrix metalloproteinase (MMP) inhibitors. Serotonin (5-HT) also increased GDNF production through FGFR2 (Tsuchioka, M., Takebayashi, M., Hisaoka, K., Maeda, N., and Nakata, Y. (2008) J. Neurochem. 106, 244-257); however, the effect of 5-HT was not inhibited by heparin and MMP inhibitors. These results suggest that amitriptyline-induced FGFR activation might occur through an extracellular pathway, in contrast to that of 5-HT. The current data show that amitriptyline-induced FGFR activation might occur by the MMP-dependent shedding of FGFR ligands, such as FGF-2, thus resulting in GDNF production.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neuroglia/cytology , Receptors, Fibroblast Growth Factor/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Humans , Mice , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Serotonin/pharmacology , Signal Transduction
6.
Brain Res ; 1384: 1-8, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21295555

ABSTRACT

Riluzole is approved for the treatment of amyotrophic lateral sclerosis (ALS); however, recent accumulating evidence suggests that riluzole is also effective for the treatment of psychiatric disorders, such as mood disorders. Plastic change in the brain induced by neurotrophic factors/growth factors is thought to be involved in the mechanism of antidepressants. This study investigated the mechanism of riluzole-induced glial cell line-derived neurotrophic factor (GDNF) production in rat C6 glioma cells (C6 cells), a model of astrocytes. The study investigated the phosphorylation of cAMP response element binding protein (CREB), an important transcriptional factor of the gdnf gene, and found that riluzole increased CREB phosphorylation in a time-dependent manner, peaking at 40min after treatment. The riluzole-induced CREB phosphorylation was completely blocked by a mitogen-activated protein kinase kinase (MEK) inhibitor (U0126). Riluzole increased extracellular signal-regulated kinase (ERK) activation prior to CREB phosphorylation. These results suggest that riluzole rapidly activates the MEK/ERK/CREB pathway. Furthermore, two types of fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitors (SU5402 and PD173074) completely blocked riluzole-induced CREB phosphorylation. In addition, riluzole rapidly phosphorylated FGFR substrate 2α (FRS2α), a major adaptor protein of FGFR. These findings suggest that riluzole induces CREB phosphorylation through FGFR. In addition, PD173074 inhibited riluzole-induced GDNF production. In contrast, l-glutamate and a glutamate transporter inhibitor (t-PDC) did not yield any effects in either CREB phosphorylation or GDNF production. These findings suggest that riluzole rapidly activates a MEK/ERK/CREB pathway through FGFR in a glutamate transporter-independent manner, followed by GDNF expression in C6 cells.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Riluzole/pharmacology , Signal Transduction/drug effects , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glioma/pathology , Glutamic Acid/pharmacology , RNA, Messenger/metabolism , Rats
7.
Article in Japanese | MEDLINE | ID: mdl-19663258

ABSTRACT

Transactivation represents the process whereby G-protein coupled receptors (GPCRs) activate receptor tyrosine kinases (RTKs), which are receptors for several neurotrophic factors and growth factors, with a subsequent downstream signaling such as mitogen-activated protein kinase (MAPK). Transactivation has been shown to be involved in the pathophysiology of cardiac hypertrophy and chronic kidney diseases, and it is targeted to develop novel therapeutic agents for these diseases. Recent accumulating evidence indicates that monoamines as well as neurotrophic factors and growth factors are thought to be involved in the therapeutic effects for psychiatric disorders. We have reported that serotonin (5-HT), which is important for the effect of antidepressant, increases glial cell line-derived neurotrophic factor (GDNF) via the 5-HT2R-mediated fibroblast growth factor receptor 2 transactivation pathway in glial cells (Tsuchioka et al, 2008). Transactivation in the CNS may play a role in the crosstalk between receptors for neurotransmitters, such as monoamines, and RTKs. Thus, transactivation will be an important basis for producing novel treatment strategies for psychiatric disorders. In this review, we outline a role of transacitvation in the CNS by explaining the concept and the mechanism of transactivation and summarizing recent reports about it in the CNS with our results.


Subject(s)
Central Nervous System/physiology , Signal Transduction/physiology , Transcriptional Activation/physiology , Animals , Receptors, CCR/physiology , Receptors, Fibroblast Growth Factor/physiology
8.
J Neurochem ; 106(1): 244-57, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18363829

ABSTRACT

We previously reported that serotonin (5-HT) increased glial cell line-derived neurotrophic factor (GDNF) release in a 5-HT(2) receptor (5-HT(2)R) and mitogen-activated protein kinase kinase/extracellular signal-related kinase (MEK/ERK)-dependent manner in rat C6 glioma cells (C6 cells), a model of astrocytes. We herein found that 5-HT-induced rapid ERK phosphorylation was blocked by 5-HT(2)R antagonists in C6 cells. We therefore examined 5-HT-induced ERK phosphorylation to reveal the mechanism of 5-HT-induced GDNF mRNA expression. As 5-HT-induced ERK phosphorylation was blocked by inhibitors for Galpha(q/11) and fibroblast growth factor receptor (FGFR), but not for second messengers downstream of Galpha(q/11), 5-HT(2)R-mediated FGFR transactivation was suggested to be involved in the ERK phosphorylation. Although FGFR1 and 2 were functionally expressed in C6 cells, 5-HT selectively phosphorylated FGFR2. Indeed, small interfering RNA for FGFR2, but not for FGFR1, blocked 5-HT-induced ERK phosphorylation. As Src family tyrosine kinase inhibitors and microtubule depolymerizing agents blocked 5-HT-induced FGFR2 phosphorylation, Src family tyrosine kinase and stabilized microtubules were suggested to act upstream of FGFR2. Finally, 5-HT-induced GDNF mRNA expression was also inhibited by the blockade of 5-HT(2)R, FGFR, and Src family tyrosine kinase. In conclusion, our findings suggest that 5-HT induces GDNF mRNA expression via 5-HT(2)R-mediated FGFR2 transactivation in C6 cells.


Subject(s)
Astrocytes/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Serotonin/metabolism , Transcriptional Activation/genetics , Animals , Astrocytes/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Microtubules/drug effects , Microtubules/metabolism , Phosphorylation/drug effects , RNA, Messenger/drug effects , RNA, Small Interfering/genetics , Rats , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin/pharmacology , Serotonin 5-HT2 Receptor Antagonists , Transcriptional Activation/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
9.
Brain Res ; 1196: 53-8, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18234163

ABSTRACT

Recently, the changes of neuronal and glial plasticity related gene expression following the increase of monoamine are suggested to be important for the therapeutic effect of antidepressants. We previously showed that antidepressants increased glial cell line-derived neurotrophic factor (GDNF) expression, which was dependent on acute activation of protein tyrosine kinase (PTK) and extracellular signal-regulated kinase (ERK) in rat C6 glioma cells (C6 cells) and normal human astrocytes (NHA). Transcription of many genes including GDNF is directed by the cAMP responsive element (CRE) and its cognate transcription factor CRE binding protein (CREB). In this study, we showed that amitriptyline, a tricyclic antidepressant, acutely increased phosphorylation of CREB, without altering the level of total CREB in C6 cells as well as in NHA. In contrast, acute amitriptyline treatment did not affect phosphorylation of CREB in SH-SY5Y cells, a human neuroblastoma cell line. Different classes of antidepressants as well as amitriptyline acutely increased phosphorylation of CREB, but haloperidol and diazepam did not. The amitriptyline-induced phosphorylation of CREB was completely blocked by U0126 [a mitogen-activated protein (MAP) kinase kinase 1 inhibitor] and genistein (a PTK inhibitor), but not by inhibitors of protein kinase A, p38 MAP kinase, or Ca(2+)/calmodulin-dependent kinase. Amitriptyline treatment also increased the expression of luciferase reporter gene regulated by CRE elements. The amitriptyline-induced luciferase activity was completely inhibited by U0126 in the same as phosphorylation of CREB. These results suggest that antidepressants acutely increase CREB activity in PTK and ERK-dependent manners, which might contribute to gene expression including GDNF in glial cells.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , CREB-Binding Protein/metabolism , Gene Expression Regulation/drug effects , Neuroglia/drug effects , Analysis of Variance , Animals , Cell Line, Tumor , Colforsin/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glioma/pathology , Humans , Phosphorylation/drug effects , Rats , Time Factors , Transfection/methods
10.
Synapse ; 62(4): 259-67, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18236460

ABSTRACT

Numerous studies have demonstrated that early adverse experiences are associated with the development of susceptibility to stress later in life. Although it is known that early experience of adversity, such as neonatal isolation, maternal separation, and low maternal care, enhances the activity of the hypothalamo-pituitary-adrenalaxis in rodents, the detailed mechanism underlying stress susceptibility induced by early adversity remains to be elucidated. Since neurotrophins have been shown to have a neuroprotective effect, we examined the influence of repeated neonatal isolation on expression of nerve growth factor (NGF), glia cell-derived neurotrophic factor (GDNF), and neurotrophin-3 mRNA in the hippocampus of juvenile and adult rats subsequently exposed immobilization stress, using real-time quantitative PCR and in situ hybridization. Neonatal isolation did not affect the basal hippocampal expression of these neurotrophin mRNAs in either juvenile or adult rats not subsequently exposed to immobilization. Similarly, there was a significant interaction between neonatal isolation and immobilization that affected the expression of NGF and GDNF mRNAs. Neonatal isolation attenuated the induction of NGF mRNA in both groups of rats and decreased GDNF mRNA in juvenile rats in response to immobilization. The decreased induction of NGF mRNA and reduced GDNF mRNA in response to immobilization was found in the CA3 pyramidal cell layer and dentate gyrus granular cell layer in the hippocampus of adult rats that had been subjected to neonatal isolation. These findings suggest that susceptibility to stress arising from prior neonatal isolation might be a result of decreased neuroprotective support through NGF and GDNF.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Hippocampus/metabolism , Nerve Growth Factor/biosynthesis , Social Isolation , Stress, Psychological/physiopathology , Animals , Animals, Newborn , Gene Expression , Hippocampus/growth & development , In Situ Hybridization , Neurotrophin 3/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Restraint, Physical , Reverse Transcriptase Polymerase Chain Reaction
11.
Nihon Shinkei Seishin Yakurigaku Zasshi ; 27(5-6): 173-9, 2007 Nov.
Article in Japanese | MEDLINE | ID: mdl-18154038

ABSTRACT

Recently, clinical and animal studies have shown that neuronal and glial plasticity are important for the therapeutic action of antidepressants. Thus, it has been suggested that neurotrophic factors or growth factors, which are potent regulators for neuronal and glial plasticity, might be involved in the effect of antidepressants. Post-mortem studies provide evidence for glial reduction in different brain areas in mood disorders. Therefore, we focused on glial cell line-derived neurotrophic factor (GDNF) in mood disorders, because GDNF plays an important role in neurogenesis and high-ordered brain function, such as learning and memory. GDNF family ligands have shown promise of efficacy for neurodegenerative disorders such as Parkinson's disease, suggesting that GDNF family ligands exist in the closest position to clinical development for treatment of diseases of the central nervous system. We reported that total GDNF levels in whole blood in patients with mood disorders were significantly lower than those in healthy control subjects (Takebayashi et al, 2006), and antidepressants increased GDNF production through monoamine-independent activation of protein tyrosine kinase (PTK) and extracellular signal-regulated kinase (ERK) in glial cells (Hisaoka et al, 2007). Clarifying the monoamine-independent novel target of antidepressants in glia might contribute to the development of more efficient therapeutics for depression.


Subject(s)
Antidepressive Agents , Glial Cell Line-Derived Neurotrophic Factor , Neuroglia , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Drug Design , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/physiology , Humans , Mood Disorders/metabolism , Mood Disorders/pathology , Neuroglia/enzymology , Neuroglia/pathology , Neuroglia/physiology , Neuronal Plasticity , Protein-Tyrosine Kinases/metabolism , Serotonin/physiology , Stimulation, Chemical
12.
J Pharmacol Exp Ther ; 321(1): 148-57, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17210798

ABSTRACT

Recent studies show that neuronal and glial plasticity are important for therapeutic action of antidepressants. We previously reported that antidepressants increase glial cell line-derived neurotrophic factor (GDNF) production in rat C6 glioma cells (C6 cells). Here, we found that amitriptyline, a tricyclic antidepressant, increased both GDNF mRNA expression and release, which were selectively and completely inhibited by mitogen-activated protein kinase kinase inhibitors. Indeed, treatment of amitriptyline rapidly increased extracellular signal-regulated kinase (ERK) activity, as well as p38 mitogen-activated protein kinase and c-Jun NH2-terminal kinase activities. Furthermore, different classes of antidepressants also rapidly increased ERK activity. The extent of acute ERK activation and GDNF release were significantly correlated to each other in individual antidepressants, suggesting an important role of acute ERK activation in GDNF production. Furthermore, antidepressants increased the acute ERK activation and GDNF mRNA expression in normal human astrocytes as well as C6 cells. Although 5-hydroxytryptamine (serotonin) (5-HT), but not noradrenaline or dopamine, increased ERK activation and GDNF release via 5-HT2A receptors, ketanserin, a 5-HT2A receptor antagonist, did not have any effect on the amitriptyline-induced ERK activation. Thus, GDNF production by amitriptyline was independent of monoamine. Both of the amitriptyline-induced ERK activation and GDNF mRNA expression were blocked by genistein, a general protein tyrosine kinase (PTK) inhibitor. Actually, we found that amitriptyline acutely increased phosphorylation levels of several phosphotyrosine-containing proteins. Taken together, these findings indicate that ERK activation through PTK regulates antidepressant-induced GDNF production and that the GDNF production in glial cells may be a novel action of the antidepressant, which is independent of monoamine.


Subject(s)
Antidepressive Agents/pharmacology , Biogenic Monoamines/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Neuroglia/enzymology , Protein-Tyrosine Kinases/metabolism , Amitriptyline/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Blotting, Western , Cell Line , Enzyme Activation/physiology , Enzyme-Linked Immunosorbent Assay , Humans , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism
13.
Int J Neuropsychopharmacol ; 9(5): 607-12, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16191208

ABSTRACT

Recent post-mortem and imaging studies provide evidence for a glial reduction in different brain areas in mood disorders. This study was aimed to test whether glial cell line-derived neurotrophic factor (GDNF), a member of transforming growth factor (TGF)-beta superfamily, in blood levels was associated with mood disorders. We measured GDNF and TGF-beta levels in whole blood in remitted patients with mood disorders [n=56; major depressive disorders (MDD) 39, bipolar disorders (BD) 17] and control subjects (n=56). GDNF and TGF-beta were assayed with the sandwich ELISA method. Total GDNF levels were significantly lower in MDD and in BD than in control subjects (MDD, p=0.0003; BD, p=0.018), while no significant difference in total TGF-beta1 or total TGF-beta2 levels was found in these groups. Our study suggests that lower GDNF levels might be involved in the pathophysiology of mood disorders, although this preliminary study has several limitations.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/blood , Mood Disorders/blood , Adult , Aged , Aged, 80 and over , Analysis of Variance , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Antimanic Agents/pharmacology , Antimanic Agents/therapeutic use , Female , Humans , Lithium Chloride/pharmacology , Lithium Chloride/therapeutic use , Male , Middle Aged , Mood Disorders/classification , Mood Disorders/drug therapy , Transforming Growth Factor beta/blood
14.
Article in Japanese | MEDLINE | ID: mdl-15796067

ABSTRACT

Recent studies show that neuronal and glial plasticity are important for the therapeutic action of antidepressants. Here, we demonstrated that amitriptyline, a tricyclic antidepressant, significantly increased GDNF mRNA and GDNF release in C6 cells. Furthermore, different classes of antidepressants increased GDNF release, but non-antidepressant psychotropic drugs did not. The amitriptyline-induced GDNF release was completely inhibited by U0126, a mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor, but was not inhibited by H-89, a protein kinase A inhibitor or calphostin C, a protein kinase C inhibitor. These results suggest that the amitriptyline-induced GDNF release may be regulated through a MEK/MAPK pathway. Next, we examined the effects of monoamines on GDNF release, because antidepressants are known to increase monoamines. 5-HT increased GDNF mRNA and GDNF release, but noradrenaline and dopamine did not. The 5-HT-induced GDNF release was partially, but significantly, blocked by ketanserin, a 5-HT2A receptor antagonist. The 5-HT-induced GDNF release was completely inhibited by U0126, but was not inhibited by H-89 or calphostin C. These results suggest that the 5-HT-induced GDNF release was mediated through a MEK/MAPK pathway and, at least, 5-HT2A receptors. GDNF, as well as other neurotrophic factors, may contribute to explain the therapeutic action of antidepressants and suggest a novel strategy of pharmacological intervention.


Subject(s)
Antidepressive Agents/pharmacology , Nerve Growth Factors/analysis , Serotonin/pharmacology , Amitriptyline/pharmacology , Animals , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Glial Cell Line-Derived Neurotrophic Factor , Isoquinolines/pharmacology , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nitriles/pharmacology , RNA, Messenger/analysis , Sulfonamides/pharmacology
15.
Brain Res ; 1002(1-2): 167-70, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-14988048

ABSTRACT

Antidepressants, which increase monoamine levels, induce glial cell line-derived neurotrophic factor (GDNF) release in C6 cells. Thus, we examined whether monoamines affect on GDNF release in C6 cells. We found that serotonin (5-HT) specifically increased GDNF mRNA expression and GDNF release in a dose- and time-dependent manner. The 5-HT-induced GDNF release was mediated through the MEK/mitogen-activated protein kinase (MAPK) pathway and, at least, 5-HT(2A) receptors. The action of 5-HT on GDNF release may provide important insights into the mechanism of antidepressants.


Subject(s)
Glioblastoma/metabolism , Nerve Growth Factors/metabolism , Serotonin/pharmacology , Animals , Cell Line, Tumor , Glial Cell Line-Derived Neurotrophic Factor , Nerve Growth Factors/biosynthesis , Rats
16.
Int Immunopharmacol ; 2(12): 1619-26, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12469936

ABSTRACT

This article reviews recent developments in cytokine research that pertain to pharmacological treatment of mood disorders such as antidepressants and lithium. We review the possible involvement of cytokines in mood disorders and their role in the therapeutic effects of antidepressant drugs. Growing evidence suggests that specific cytokines signal the brain to generate neurochemical, neuroimmune, neuroendocrine and behavior changes. An imbalance of cytokines within the central nervous system (CNS), or even systemically, may play a role in the pathophysiology of mood disorders. Modulation of these cytokines by chronic antidepressant treatment may result in restored balance. However, the effect of antidepressants on cytokines is still unclear both in clinical and preclinical research due to limited data. Further research is needed to clarify the involvement of cytokines in mood disorders. Understanding this relationship may lead to rational, therapeutic improvements in antidepressant and mood stabilizing drugs.


Subject(s)
Antidepressive Agents/therapeutic use , Cytokines/metabolism , Mood Disorders/drug therapy , Mood Disorders/metabolism , Animals , Antidepressive Agents/pharmacology , Humans , Immune System/drug effects , Lithium/pharmacology , Monoamine Oxidase Inhibitors/pharmacology
17.
Jpn J Pharmacol ; 89(3): 242-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12184729

ABSTRACT

We investigated the mechanisms of 5-HT-induced tachycardia, which we reported previously to be triggered by 5-HT3 receptor stimulation, in the isolated guinea pig atrium in comparison with that induced by isoproterenol and histamine. We found that 5-HT-induced tachycardia was completely inhibited by ruthenium red. 5-HT-induced tachycardia was reduced in the capsaicin pre-treated atrium as well as in the presence of capsaicin. The effects of isoproterenol and histamine were not affected by ruthenium red or capsaicin treatment. Furthermore, 5-HT-induced tachycardia was found to be potentiated by thiorphan, an inhibitor of peptide degeneration. Calcitonin gene-related peptide (CGRP) (1-37), a full agonist of CGRP1-like receptors, was found to act selectively as a potent stimulator of chronotropic action. CGRP (8-37), an antagonist of CGRP1-type receptors, inhibited 5-HT-induced tachycardia as well as effects induced by CGRP (1-37). The observation that tetrodotoxin failed to affect 5-HT-induced tachycardia excluded the involvement of 5-hydroxytryptaminergic interneurons. Thus, we confirmed that the mechanism of 5-HT-induced tachycardia is distinct from that induced by isoproterenol and histamine. In conclusion, the activation of 5-HT3 receptors on the sensory nerve terminals brought about ruthenium red-sensitive Ca2+ influx and resulted in the release of CGRP from capsaicin-sensitive stores, and then CGRP stimulated CGRP1-like receptors to produce 5-HT-induced tachycardia.


Subject(s)
Capsaicin/pharmacology , Heart Rate/drug effects , Receptors, Serotonin/physiology , Ruthenium Red/pharmacology , Serotonin/pharmacology , Animals , Dose-Response Relationship, Drug , Guinea Pigs , Heart Atria/drug effects , Heart Rate/physiology , In Vitro Techniques , Receptors, Serotonin, 5-HT3 , Stimulation, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL
...