Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Psychiatry Neurosci ; 43(5): 338-346, 2018 08.
Article in English | MEDLINE | ID: mdl-30125244

ABSTRACT

BACKGROUND: Results of neuroimaging and postmortem studies suggest that people with schizophrenia may have lower levels of muscarinic M1 receptors (CHRM1) in the cortex, but not in the hippocampus or thalamus. Here, we use a novel immunohistochemical approach to better understand the likely cause of these low receptor levels. METHODS: We determined the distribution and number of CHRM1-positive (CHRM1+) neurons in the cortex, medial dorsal nucleus of the thalamus and regions of the hippocampus from controls (n = 12, 12 and 5, respectively) and people with schizophrenia (n = 24, 24 and 13, respectively). RESULTS: Compared with controls, levels of CHRM1+ neurons in people with schizophrenia were lower on pyramidal cells in layer III of Brodmann areas 9 (-44%) and 17 (-45%), and in layer V in Brodmann areas 9 (-45%) and 17 (-62%). We found no significant differences in the number of CHRM1+ neurons in the medial dorsal nucleus of the thalamus or in the hippocampus. LIMITATIONS: Although diagnostic cohort sizes were typical for this type of study, they were relatively small. As well, people with schizophrenia were treated with antipsychotic drugs before death. CONCLUSION: The loss of CHRM1+ pyramidal cells in the cortex of people with schizophrenia may underpin derangements in the cholinergic regulation of GABAergic activity in cortical layer III and in cortical/subcortical communication via pyramidal cells in layer V.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Mediodorsal Thalamic Nucleus/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Receptor, Muscarinic M1/metabolism , Schizophrenia/metabolism , Adult , Autopsy , Brain/cytology , Brain/metabolism , Case-Control Studies , Cell Count , Cerebral Cortex/cytology , Female , Hippocampus/cytology , Humans , Immunohistochemistry , Male , Mediodorsal Thalamic Nucleus/cytology , Middle Aged , Neurons/cytology , Pyramidal Cells/cytology , Schizophrenia/pathology
2.
J Psychiatry Neurosci ; 43(4): 170202, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29848411

ABSTRACT

BACKGROUND: Results of neuroimaging and postmortem studies suggest that people with schizophrenia may have lower levels of muscarinic M1 receptors (CHRM1) in the cortex, but not in the hippocampus or thalamus. Here, we use a novel immunohistochemical approach to better understand the likely cause of these low receptor levels. METHODS: We determined the distribution and number of CHRM1-positive (CHRM1+) neurons in the cortex, medial dorsal nucleus of the thalamus and regions of the hippocampus from controls (n = 12, 12 and 5, respectively) and people with schizophrenia (n = 24, 24 and 13, respectively). RESULTS: Compared with controls, levels of CHRM1+ neurons in people with schizophrenia were lower on pyramidal cells in layer III of Brodmann areas 9 (-44%) and 17 (-45%), and in layer V in Brodmann areas 9 (-45%) and 17 (-62%). We found no significant differences in the number of CHRM1+ neurons in the medial dorsal nucleus of the thalamus or in the hippocampus. LIMITATIONS: Although diagnostic cohort sizes were typical for this type of study, they were relatively small. As well, people with schizophrenia were treated with antipsychotic drugs before death. CONCLUSION: The loss of CHRM1+ pyramidal cells in the cortex of people with schizophrenia may underpin derangements in the cholinergic regulation of GABAergic activity in cortical layer III and in cortical/subcortical communication via pyramidal cells in layer V.

3.
J Chem Neuroanat ; 77: 187-192, 2016 11.
Article in English | MEDLINE | ID: mdl-27435807

ABSTRACT

The muscarinic M1 receptor plays a significant role in cognition, probably by modulating information processing in key regions such as the hippocampus. To understand how the muscarinic M1 receptor achieves these functions in the hippocampus, it is critical to know the distribution of the receptor within this complex brain region. To date, there are limited data on the distribution of muscarinic M1 receptors in the human hippocampus which may also be confounded because some anti-muscarinic receptor antibodies have been shown to lack specificity. Initially, using Western blotting and immunohistochemistry, we showed the anti-muscarinic M1 receptor antibody to be used in our study bound to a single 62kDa protein that was absent in mice lacking the muscarinic M1 receptor gene. Then, using immunohistochemistry, we determined the distribution of muscarinic M1 receptors in human hippocampus from 10 subjects with no discernible history of a neurological or psychiatric disorder. Our data shows the muscarinic M1 receptor to be predominantly on pyramidal cells in the hippocampus. Muscarinic M1 receptor positive cells were most apparent in the deep polymorphic layer of the dentate gyrus, the pyramidal cell layer of cornu ammonis region 3, the cellular layers of the subiculum, layer II of the presubiculum and layer III and V of the parahippocampal gyrus. Positive cells were less numerous and less intensely stained in the pyramidal layer of cornu ammonis region 2 and were sparse in the molecular layer of the dentate gyrus as well as cornu ammonis region 1. Although immunoreactivity was present in the granular layer of the dentate gyrus, it was difficult to identity individual immunopositive cells, possibly due to the density of cells. This distribution of the muscarinic M1 receptors in human hippocampus, and its localisation on glutamatergic cells, would suggest the receptor has a significant role in modulating excitatory hippocampal neurotransmission.


Subject(s)
Hippocampus/metabolism , Receptor, Muscarinic M1/metabolism , Adult , Aged , Animals , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Female , Hippocampus/anatomy & histology , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Middle Aged , Parahippocampal Gyrus/cytology , Parahippocampal Gyrus/metabolism , Pyramidal Cells/metabolism , Reference Values , Synaptic Transmission , Young Adult
4.
Clin Psychopharmacol Neurosci ; 12(2): 94-110, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25191500

ABSTRACT

Psychiatric disorders are among the most debilitating of all medical illnesses. Whilst there are drugs that can be used to treat these disorders, they give sub-optimal recovery in many people and a significant number of individuals do not respond to any treatments and remain treatment resistant. Surprisingly, the mechanism by which psychotropic drugs cause their therapeutic benefits remain unknown but likely involves the underlying molecular pathways affected by the drugs. Hence, in this review, we have focused on recent findings on the molecular mechanism affected by antipsychotic, mood stabilizing and antidepressant drugs at the levels of epigenetics, intracellular signalling cascades and microRNAs. We posit that understanding these important interactions will result in a better understanding of how these drugs act which in turn may aid in considering how to develop drugs with better efficacy or increased therapeutic reach.

5.
Schizophr Res ; 158(1-3): 247-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25037527

ABSTRACT

We previously identified a group of subjects with schizophrenia who, on average, have a 75% decrease in cholinergic receptor, muscarinic 1 (CHRM1) in Brodmann's area (BA) 9. To extend this finding, we determined i) if the decrease in CHRM1 was present in another functionally related CNS region (BA6), ii) whether the marked decrease in CHRM1 was accompanied by changes in levels of other CHRMs and iii) potential factors responsible for the decreased CHRM1 expression. We measured CHRM1 and CHRM3 using in situ radioligand binding with [(3)H]pirenzepine and [(3)H]4-DAMP respectively in BA6 from 20 subjects with schizophrenia who had low levels of CHRM1 in BA9 (SzLow[(3)H]PZP), 18 subjects with schizophrenia whose levels of CHRM1 were similar to controls (SzNormal[(3)H]PZP) and 20 control subjects. Levels of CHRM1, 3 and 4 mRNA were measured using qPCR and levels of the transcription factors, SP1 and SP3, were determined using Western blots. In BA6, the density of [(3)H]pirenzepine binding was decreased in subjects with SzLow[(3)H]PZP (p<0.001) compared to controls. The density of [(3)H]4-DAMP binding, levels of CHRM1, 3 and 4 mRNA and levels of SP1 and SP3 was not significantly different between the three groups. This study shows that the previously identified decrease in CHRM1 expression is not confined to the dorsolateral prefrontal cortex but is present in other cortical areas. The effect shows some specificity to CHRM1, with no change in levels of binding to CHRM3. Furthermore, this decrease in CHRM1 does not appear to be associated with low levels of CHRM1 mRNA or to simply be regulated by the transcription factors, SP1 and SP3, suggesting that other mechanisms are responsible for the decreased CHRM1 in these subjects.


Subject(s)
Receptors, Muscarinic/metabolism , Schizophrenia/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Adult , Blotting, Western , Brain/metabolism , Cohort Studies , Humans , Middle Aged , Piperidines , Pirenzepine , Polymerase Chain Reaction , RNA, Messenger/metabolism , Radioligand Assay , Receptor, Muscarinic M1 , Receptor, Muscarinic M3 , Receptor, Muscarinic M4 , Tritium
6.
Int J Neuropsychopharmacol ; 17(6): 895-906, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24495390

ABSTRACT

We have attempted to replicate studies showing higher levels of serotonin 2A receptors (HTR2A) in the cortex of people with mood disorders and to determine the effects of treating rats with antidepressant drugs on levels of that receptor. In situ [3H]ketanserin binding and autoradiography was used to measure levels of HTR2A in Brodmann's area (BA) 46 and 24 from people with major depressive disorders (MDD, n = 16), bipolar disorders (BD, n = 14) and healthy controls (n = 14) as well as the central nervous system (CNS) of rats (20 per treatment arm) treated for 10 or 28 d with fluoxetine (10 mg/kg/d) or imipramine (20 mg/kg/d). Compared with controls, HTR2A were lower in BA 24, but not BA 46, from people with MDD (p = 0.005); HTR2A were not changed in BD. Levels of HTR2A were lower in BA 24 (p = 0.007), but not BA 46, from people who had died by suicide. Finally, levels of HTR2A were lower in the CNS of rats treated with imipramine, but not fluoxetine, for 28 d, but not 10 d. From our current and previous data we conclude cortical HTR2A are lower in schizophrenia, MDD, people with mood disorders who died by suicide, rats treated with some antipsychotic or some antidepressant drugs. As levels of cortical HTR2A can be affected by the aetiologies of different disorders and mechanisms of action of different drugs, a better understanding of how such changes can occur needs to be elucidated.


Subject(s)
Antidepressive Agents/pharmacology , Cerebral Cortex/drug effects , Depressive Disorder, Major/drug therapy , Imipramine/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Suicide , Adult , Aged , Aged, 80 and over , Animals , Antipsychotic Agents/pharmacology , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Cerebral Cortex/metabolism , Depressive Disorder, Major/metabolism , Female , Fluoxetine/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Humans , Male , Middle Aged , Rats, Sprague-Dawley , Young Adult
7.
Psychopharmacology (Berl) ; 230(3): 425-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23771439

ABSTRACT

RATIONALE: Clozapine affects the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway in the brain, which plays an important role in its antipsychotic action. However, previous findings are inconsistent, and related molecular mechanisms require further clarification. OBJECTIVES: Time- and dose-dependent effects of clozapine on the ERK1/2 pathway and its regulatory mechanism were investigated in rat frontal cortex. METHODS AND RESULTS: At 15, 30, 60, and 120 min after intraperitoneal injection of clozapine (5, 10, and 20 mg/kg), changes in ERK1/2, its upstream canonical kinases (Raf1 and mitogen-activated protein kinase kinase 1/2 [MEK1/2]), and its downstream molecule (p90 ribosomal S6 kinase [p90RSK]) were investigated in rat frontal cortex. At 15 min, p-Raf1, p-MEK1/2, p-ERK1/2, and p-p90RSK all increased dose-dependently. At 30 min, p-ERK1/2 and p-p90RSK showed no significant changes, while dose-dependent increases in p-Raf1 and p-MEK1/2 were found. At 60 and 120 min, although p-ERK1/2 and p-p90RSK decreased, increases in p-Raf1 and p-MEK1/2 were maintained. A clozapine-induced reduction in ERK1/2 phosphorylation was evident at both tyrosine and threonine residues, suggesting the involvement of dual specificity phosphatases (DUSPs; mitogen-activated protein kinase phosphatases [MKPs]). mRNA expression of seven Dusps that can dephosphorylate ERK1/2 were examined; Mkp-1 (Dusp1) mRNA increased following clozapine treatment. Moreover, MKP-1 protein and phosphatase activity increased, and binding of MKP-1 to ERK1/2 was also upregulated by clozapine administration. CONCLUSIONS: In rat frontal cortex, clozapine regulates ERK1/2 phosphorylation via MKP-1, which induces uncoupling between Raf1-MEK1/2 and ERK1/2-p90RSK activity. These findings suggest an important role of MKP-1 in the mechanism of action of clozapine.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Dual Specificity Phosphatase 1/metabolism , Animals , Antipsychotic Agents/administration & dosage , Clozapine/administration & dosage , Dose-Response Relationship, Drug , Frontal Lobe/drug effects , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-raf , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Time Factors
8.
Front Biosci (Landmark Ed) ; 16(3): 962-79, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196212

ABSTRACT

APOE is a major component of several lipoproteins. In addition to its role as a lipid transport protein APOE also serves a dual role as a glial derived, synaptic signalling molecule and thought to play an important role in synaptic plasticity and cognition. Polymorphisms within the APOE gene have been associated with the incidence of Alzheimer's disease. In light of the similarities in the cognitive deficits experienced in both Alzheimer's disease and schizophrenia as well as the comorbidity of depression in Alzheimer's disease, aberrant APOE signalling has been implicated in the pathologies of schizophrenia and mood disorders. The schizophrenia candidate gene, reelin, also shares common receptors with APOE, further supporting a role for APOE in the pathology of these disorders. This review will summarise the current understanding of the involvement of APOE and its receptors in the symptomatology and pathology of schizophrenia and mood disorders and the implications of this involvement for drug treatment.


Subject(s)
Apolipoproteins E/genetics , Apolipoproteins E/physiology , Mood Disorders/genetics , Schizophrenia/genetics , Alzheimer Disease/genetics , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Cell Adhesion Molecules, Neuronal/physiology , Central Nervous System/metabolism , Cognition/physiology , Depressive Disorder, Major/physiopathology , Extracellular Matrix Proteins/physiology , Humans , Lipid Metabolism , Mood Disorders/physiopathology , Myelin Sheath/pathology , Nerve Tissue Proteins/physiology , Neuroglia/physiology , Neuronal Plasticity/drug effects , Receptors, LDL/physiology , Reelin Protein , Schizophrenia/physiopathology , Serine Endopeptidases/physiology , Signal Transduction/physiology , Synapses/physiology
9.
Psychiatry Res ; 167(1-2): 80-7, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19342105

ABSTRACT

Repeated administration of NMDA antagonists can induce behavioral alterations that mimic symptoms of psychosis, as seen in schizophrenia. JNK, one of the MAPKs, and c-Jun, its downstream target molecule, play important roles in regulating apoptosis in neural cells, and have been suggested as being associated with the pathophysiology of psychosis and the mechanism of action of some antipsychotics. We investigated changes in the JNK-c-Jun pathway and other Jun family proteins in the rat frontal cortex after single and repeated administration of MK-801 to examine acute and chronic responses. Neither the protein level nor the phosphorylation of JNK changed after single or repeated doses of MK-801. However, after repeated treatments, but not a single treatment, with MK-801, a down-regulation occurred in the protein level and of Ser73 phosphorylation of c-Jun in the rat frontal cortex. Other members of the Jun family, JunB and JunD, were unchanged. Repeated exposure to MK-801 down-regulated the phosphorylation and protein level of c-Jun in the rat frontal cortex, which may be related to the long-term effects of chronic treatment with MK-801.


Subject(s)
Dizocilpine Maleate/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Frontal Lobe/metabolism , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Antipsychotic Agents/pharmacology , Apoptosis/drug effects , Behavior, Animal/drug effects , Down-Regulation/drug effects , Frontal Lobe/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/chemically induced , Schizophrenia/metabolism , Signal Transduction/drug effects
10.
Exp Mol Med ; 40(4): 435-44, 2008 Aug 31.
Article in English | MEDLINE | ID: mdl-18779656

ABSTRACT

Repeated electroconvulsive seizure (ECS), a model for electroconvulsive therapy (ECT), exerts neuroprotective and proliferative effects in the brain. This trophic action of ECS requires inhibition of apoptotic activity, in addition to activation of survival signals. c-Myc plays an important role in apoptosis of neurons, in cooperation with the Bcl-2 family proteins, and its activity and stability are regulated by phosphorylation and ubiquitination. We examined c-Myc and related proteins responsible for apoptosis after repeated ECS. In the rat frontal cortex, repeated ECS for 10 days reduced the total amount of c-Myc, while increasing phosphorylation of c-Myc at Thr58, which reportedly induces degradation of c-Myc. As expected, ubiquitination of both phosphorylated and total c-Myc increased after 10 days ECS, suggesting that ECS may reduce c-Myc protein level via ubiquitination-proteasomal degradation. Bcl-2 family proteins, caspase, and poly(ADP-ribose) polymerase (PARP) were investigated to determine the consequence of down-regulating c-Myc. Protein levels of Bcl-2, Bcl-X(L), Bax, and Bad showed no change, and cleavage of caspase-3 and PARP were not induced. However, phosphorylation of Bad at Ser-155 and binding of Bad to 14-3-3 increased without binding to Bcl-X(L) after repeated ECS, implying that repeated ECS sequesters apoptotic Bad and frees pro-survival Bcl-XL. Taken together, c-Myc down-regulation via ubiquitination-proteasomal degradation and Bad inactivation by binding to 14-3-3 may be anti-apoptotic mechanisms elicited by repeated ECS in the rat frontal cortex. This finding further supports the trophic effect of ECS blocking apoptosis as a possible therapeutic effect of ECT.


Subject(s)
Electroconvulsive Therapy/adverse effects , Frontal Lobe/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Seizures/metabolism , bcl-Associated Death Protein/metabolism , 14-3-3 Proteins/metabolism , Animals , Down-Regulation , Male , Models, Biological , Neurons/metabolism , Periodicity , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Seizures/etiology , Tumor Cells, Cultured , Ubiquitination , bcl-Associated Death Protein/antagonists & inhibitors
11.
Int J Neuropsychopharmacol ; 11(4): 509-17, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18272021

ABSTRACT

Haloperidol, a classical antipsychotic drug, affects the extracellular signal-regulated kinase (ERK) pathway in the brain. However, findings are inconsistent and the mechanism by which haloperidol regulates ERK is poorly understood. Therefore, we examined the ERK pathway and the related protein phosphatase 2A (PP2A) in detail after haloperidol administration. Haloperidol (0.5 and 1 mg/kg) induced biphasic changes in the phosphorylation level of mitogen-activated protein kinase kinase (MEK), ERK, and p90 ribosomal S6 kinase (p90RSK) without changing Raf-1 phosphorylation. Fifteen minutes after haloperidol administration, MEK-ERK-p90RSK phosphorylation increased, whilst PP2A activity decreased. At 60 min, the reverse was observed and the binding of PP2A to MEK and ERK increased. Higher dosages of haloperidol (2 and 4 mg/kg), affected neither MEK-ERK-p90RSK phosphorylation nor PP2A activity. Accordingly, PP2A regulates acute dose- and time-dependent changes in MEK-ERK-p90RSK phosphorylation after haloperidol treatment. These findings suggest the involvement of a dephosphorylating mechanism in the acute action of haloperidol.


Subject(s)
Antipsychotic Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Haloperidol/pharmacology , Mitogen-Activated Protein Kinases/physiology , Prefrontal Cortex/physiology , Protein Phosphatase 2/physiology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Signal Transduction/drug effects , Animals , Blotting, Western , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/genetics , Immunoprecipitation , Male , Mitogen-Activated Protein Kinases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/physiology , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/genetics , raf Kinases/physiology
12.
Neurosci Lett ; 434(1): 23-8, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18262357

ABSTRACT

In experimental animals, including rats, MK-801 produces characteristic behavioural changes that model schizophrenia. It has been hypothesized that these changes accompany long-term synaptic changes, which require protein neosynthesis. We observed the effect of MK-801 on the "mammalian target of rapamycin" (mTOR)/70-kDa ribosomal protein S6 kinase (p70S6K) pathway that regulates protein synthesis in the rat frontal cortex. A single injection of MK-801 (0.5, 1, or 2mg/kg) induced an acute increase in the phosphorylation of Akt (Ser-473) eIF4E-binding protein (4E-BP1) (Thr-37/46) and p70S6K (Thr-389). In contrast, after repeated treatment with MK-801 (1mg/kg for 5 or 10 days), the phosphorylation of Akt (Ser-473), mTOR (Ser-2481), 4E-BP1 (Thr-37/46), p70S6K (Thr-389), and S6 (Ser-240/244) increased. Thus, proteins in the mTOR/p70S6K pathway are modulated in chronic MK-801 animal models. These findings may suggest that repeated MK-801 treatment activates the signal transduction pathways involved in the initiation of protein synthesis in the rat frontal cortex.


Subject(s)
Dizocilpine Maleate/pharmacology , Prefrontal Cortex/drug effects , Protein Biosynthesis/drug effects , Protein Kinases/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Animals , Eukaryotic Initiation Factor-4E/biosynthesis , Eukaryotic Initiation Factor-4E/drug effects , Eukaryotic Initiation Factor-4E/genetics , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Hallucinogens/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Protein Biosynthesis/genetics , Protein Kinases/biosynthesis , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
13.
Exp Mol Med ; 39(3): 353-60, 2007 Jun 30.
Article in English | MEDLINE | ID: mdl-17603289

ABSTRACT

Glycogen synthase kinase 3 (GSK3) was recently suggested to be a potential target of psychotropics used in psychiatric illnesses such as schizophrenia and bipolar disorder. Relevant studies have found that antipsychotic drugs regulate GSK3 activity via an increase in either inhibitory serine phosphorylation or amount of GSK3 after acute or subchronic treatment. Recent evidence shows that GSK3 is regulated by dopaminergic or serotonergic systems implicated in the pathophysiology and treatment mechanisms of schizophrenia and bipolar disorder. Therefore, antipsychotics may regulate GSK3 via antagonizing dopaminergic or serotonergic activity. However, the signaling pathway that is involved in GSK3 regulation by dopaminergic or serotonergic systems has not been well established. Haloperidol is a typical antipsychotic with potent dopamine D(2) receptor antagonism. Clozapine is an atypical antipsychotic with potent serotonin 5HT(2) receptor antagonism. We injected rats with haloperidol or clozapine and examined the phosphorylation and amount of GSK3alpha/beta and its well-known upstream regulators Akt and Dvl in the rat frontal cortex by Western blotting. Both haloperidol and clozapine induced Ser21/9 phosphorylation of GSK3GSK3alpha/beta. Haloperidol increased the Ser473 phosphorylation of Akt transiently, whereas clozapine maintained the increase for 1 h. Haloperidol did not affect the phosphorylation and amount of Dvl, whereas clozapine increased both phosphorylation and the amount of Dvl. Our results suggest that GSK3 activity may be regulated by both typical and atypical antipsychotics and that Akt or Dvl, depending on the D(2)- or 5HT(2)- receptor antagonism properties of typical and atypical antipsychotics, mediate the regulation differently.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Frontal Lobe/drug effects , Glycogen Synthase Kinase 3/metabolism , Haloperidol/pharmacology , Phosphoproteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dishevelled Proteins , Dopamine Antagonists/pharmacology , Frontal Lobe/enzymology , Male , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology , Signal Transduction
14.
Int J Neuropsychopharmacol ; 10(3): 359-68, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16780607

ABSTRACT

Repeated administrations of NMDA receptor antagonists induce behavioural changes which resemble the symptoms of schizophrenia in animals. ERK and GSK-3beta associated signalling pathways have been implicated in the pathogenesis of psychosis and in the action mechanisms of various psychotropic agents. Here, we observed the phosphorylations of ERK and GSK-3beta and related molecules in the rat frontal cortex after repeated intraperitoneal injections of MK-801, over periods of 1, 5, and 10 d. Repeated treatment with 0.5, 1, and 2 mg/kg MK-801 increased the phosphorylation levels of the MEK-ERK-p90RSK and Akt-GSK-3beta pathways and concomitantly and significantly increased CREB phosphorylation in the rat frontal cortex. However, single MK-801 treatment did not induce these significant changes. In addition, the immunoreactivities of HSP72, Bax, and PARP were not altered, which suggests that neuronal damage may not occur in the rat frontal cortex in response to chronic MK-801 treatment. These findings suggest that chronic exposure to MK-801 may induce pro-survival and anti-apoptotic activity without significant neuronal damage in the rat frontal cortex. Moreover, this adaptive change might be associated with the psychotomimetic action of MK-801.


Subject(s)
Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Glycogen Synthase Kinase 3/physiology , Prefrontal Cortex/drug effects , Signal Transduction/drug effects , Animals , Blotting, Western , Cyclic AMP Response Element-Binding Protein/physiology , Glycogen Synthase Kinase 3 beta , HSP72 Heat-Shock Proteins/metabolism , Immunohistochemistry , Male , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , bcl-2-Associated X Protein/biosynthesis
15.
Int J Neuropsychopharmacol ; 9(4): 451-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16079022

ABSTRACT

MK-801 induces psychotomimetic behavioural changes in animals. ERKs play an important role in the pathogenesis of schizophrenia and in the action of antipsychotics and psychotomimetics. We observed phosphorylation of ERK-signalling-pathway-associated molecules in the rat frontal cortex and their association with rat behaviour after MK-801 administration. After injecting 0.25-1 mg/kg MK-801, ERK phosphorylation decreased compared to vehicle treatment, and rats showed increased locomotion. After 2 mg/kg treatment, ERK phosphorylation increased and rat motility started to decrease. After treating with 4-8 mg/kg, ERK phosphorylation once again decreased and rats showed hypomotility and ataxia. ERK phosphorylation levels were maintained from 15 min to 90 min after 1 or 2 mg/kg treatment. Ser338-c-Raf and MEK phosphorylation showed similar dose-dependent and temporal patterns to those of ERK. Taken together, Ser338-c-Raf-MEK-ERK phosphorylation by MK-801 in the rat frontal cortex showed a specific pattern and may be associated with behavioural changes induced by MK-801.


Subject(s)
Dizocilpine Maleate/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/enzymology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
16.
Int J Mol Med ; 16(5): 841-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16211253

ABSTRACT

Sodium salicylate, one of anti-inflammatory agents, is known to partially induce the heat shock response: it stimulates the DNA-binding of heat shock factor 1 (HSF1) without inducing heat shock gene expression. Here we show that when C6 glioma cells are recovered from sodium salicylate treatment, they highly induce heat shock protein 72 (HSP72), but not HSP73 and HSP90, demonstrating that salicylate-induced inert HSF1 can be fully activated into a transcriptionally competent form by sodium salicylate recovery (SR)-specific mechanism. Fluorescent analysis using 2',7'-dichlorodihydrofluorescein diacetate revealed that sodium salicylate enhanced reactive oxygen species (ROS) production. N-acetyl-L-cysteine (NAC, a ROS scavenger) completely suppressed SR-induced HSP72 synthesis and HSP72 promoter-driven CAT reporter gene transcription as well as salicylate-induced HSF1-DNA binding, indicating a critical role(s) of ROS in the SR-induced HSP72 gene regulation. We also show that treatment of C6 cells with sodium salicylate activated p38MAPK and inactivated ERK1/2 in a ROS-independent manner and activities of these protein kinases returned during recovery period to the control level. Inhibiting p38MAPK and ERK1/2 with the p38MAPK inhibitors (SB203580 and SB202190) and the MEK1/2 inhibitor (PD98059 and U0126) or with expression of dominant negative p38MAPK and ERK1/2 abolished SR-induced HSP72 synthesis and HSP70 promoter-driven CAT activity. However, sodium salicylate-induced HSF1-DNA binding was not affected by the p38MAPK inhibitor or the MEK1/2 inhibitor. These findings suggest that sodium salicylate partially activates HSF1 via ROS production and p38MAPK activation and the salicylate-induced inert HSF1 can be fully activated into a transcriptionally competent form by the ERK1/2 signaling pathways that are activated independently of ROS during SR.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , DNA-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Sodium Salicylate/pharmacology , Transcription Factors/metabolism , Animals , DNA/metabolism , Glioblastoma , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Promoter Regions, Genetic , Rats , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Int J Neuropsychopharmacol ; 8(4): 607-13, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15877933

ABSTRACT

GSK-3beta is regarded as playing an important part in the pathogenesis of schizophrenia and the action of psychotomimetic agents. We observed phosphorylation of molecules associated with the GSK-3beta signalling pathway in the rat brain after MK-801 injection, which induces a schizophrenia-like state in humans. Ser9-GSK-3beta phosphorylation was increased after injection of 1 mg/kg MK-801 in the rat frontal cortex but not in the hippocampus or cerebellum. This increase peaked at 30 min and was maintained until 90 min after injection. The phosphorylation showed a dose-dependent increase up to 1 mg/kg MK-801, followed by a decrease at higher dosage. Furthermore, phosphorylation of Ser473-Akt and Ser133-CREB showed similar temporal, dose-dependent and regionally specific patterns with those of Ser9-GSK-3beta. However, phosphorylation of Dvl and Ser33-beta-catenin was not affected by MK-801. These results suggest that GSK-3beta phosphorylation by MK-801 may be associated with the Akt-GSK-3beta pathway rather than with the Wnt-Dvl-GSK3beta pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glycogen Synthase Kinase 3/metabolism , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Actins/metabolism , Animals , Blotting, Western , Catenins/metabolism , Dizocilpine Maleate/administration & dosage , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Glycogen Synthase Kinase 3 beta , Injections, Intraperitoneal , Male , Phosphorylation , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley
18.
Biol Psychiatry ; 57(1): 107-9, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15607308

ABSTRACT

BACKGROUND: Recent reports indicate that repeated electroconvulsive shock (ECS) induces cortical cell proliferation, suggesting the possibility that ECS may activate cell cycle progression in the rat brain cortex. METHODS: Sprague-Dawley rats (150-200g) were divided into four treatment groups and then given sham treatment or ECS treatment for 1, 5, and 10 days, respectively. The activity of cyclin-dependent kinase 2 (Cdk2), phosphorylation, and total protein amount of cyclin D1, cyclin E, pocket retinoblastoma family of protein (pRB), and E2F1 were analyzed in the rat cerebral cortex. RESULTS: The activity of Cdk2, the protein amount of pRB, Ser795 phosphorylation of pRB, and the protein amount of E2F1 were all increased compared with the sham-treated control subjects, and these increases were enhanced with the increasing number of ECS. In contrast, the protein amounts of Cdk2, cyclin D1, and cyclin E were not changed by repeated ECS. CONCLUSIONS: The Cdk2-pRB-E2F1 cell cycle pathway is activated by repeated ECS in the rat frontal cortex.


Subject(s)
CDC2-CDC28 Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/radiation effects , DNA-Binding Proteins/metabolism , Electroshock , Frontal Lobe/radiation effects , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western/methods , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Activation/radiation effects , Frontal Lobe/metabolism , Male , Phosphorylation/radiation effects , Rats , Rats, Sprague-Dawley , Serine/metabolism , Time Factors
19.
FEBS Lett ; 560(1-3): 115-9, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14988008

ABSTRACT

We investigated the effect of 10 microM clozapine on the activity of glycogen synthase kinase-3beta (GSK-3beta) and its upstream and downstream molecules in SH-SY5Y human neuroblastoma cells. Clozapine activates both Akt- and Dvl-mediated phosphorylation of GSK-3beta through phosphorylation at Ser9, and increased total cellular and intranuclear levels of beta-catenin. Pretreatment with the specific inhibitor of the phosphatidylinositol 3-kinase (PI3K)-Akt pathway, LY294002 (20 microM), prevented the phosphorylation of Akt but did not affect the phosphorylation of GSK-3beta. These results suggest that clozapine regulates the phosphorylation of GSK-3beta through Wnt signal pathways involving Dvl upstream but not through the PI3K-Akt pathway in SH-SY5Y cells.


Subject(s)
Clozapine/pharmacology , Glycogen Synthase Kinase 3/metabolism , Protein Serine-Threonine Kinases , Signal Transduction , Zebrafish Proteins , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Nucleus/chemistry , Chromones/pharmacology , Culture Media, Serum-Free , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3 beta , Humans , Morpholines/pharmacology , Neuroblastoma/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Serine/metabolism , Time Factors , Trans-Activators/analysis , Trans-Activators/drug effects , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...