Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Transl Psychiatry ; 7(2): e1025, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28170001

ABSTRACT

Post-traumatic stress disorder (PTSD) is psychiatric disease, which can occur following exposure to traumatic events. PTSD may be acute or chronic, and can have a waxing and waning course of symptoms. It has been hypothesized that proinflammatory cytokines and chemokines in the cerebrospinal fluid (CSF) or plasma might be mediators of the psychophysiological mechanisms relating a history of trauma exposure to changes in behavior and mental health disorders, and medical morbidity. Here we test the cytokine/chemokine hypothesis for PTSD by examining levels of 17 classical cytokines and chemokines in CSF, sampled at 0900 hours, and in plasma sampled hourly for 24 h. The PTSD and healthy control patients are from the NIMH Chronic PTSD and healthy control cohort, initially described by Bonne et al. (2011), in which the PTSD patients have relatively low comorbidity for major depressive disorder (MDD), drug or alcohol use. We find that in plasma, but not CSF, the bivariate MCP4 (CCL13)/ MCP1(CCL2) ratio is ca. twofold elevated in PTSD patients compared with healthy controls. The MCP-4/MCP-1 ratio is invariant over circadian time, and is independent of gender, body mass index or the age at which the trauma was suffered. By contrast, MIP-1ß is a candidate biomarker for PTSD only in females, whereas TARC is a candidate biomarker for PTSD only in males. It remains to be discovered whether these disease-specific differences in circadian expression for these specific immune signaling molecules are biomarkers, surrogates, or drivers for PTSD, or whether any of these analytes could contribute to therapy.


Subject(s)
Chemokine CCL2/metabolism , Monocyte Chemoattractant Proteins/metabolism , Stress Disorders, Post-Traumatic/metabolism , Adult , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Chemokine CCL17/metabolism , Chemokine CCL4/metabolism , Chronic Disease , Circadian Rhythm , Cytokines/metabolism , Female , Humans , Male , Sex Factors
2.
Mol Psychiatry ; 18(8): 856-63, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689537

ABSTRACT

Available treatments for depression have significant limitations, including low response rates and substantial lag times for response. Reports of rapid antidepressant effects of a number of compounds, including the glutamate N-methyl-D-aspartate receptor antagonist ketamine, have spurred renewed translational neuroscience efforts aimed at elucidating the molecular and cellular mechanisms of action that result in rapid therapeutic response. This perspective provides an overview of recent advances utilizing compounds with rapid-acting antidepressant effects, discusses potential mechanism of action and provides a framework for future research directions aimed at developing safe, efficacious antidepressants that achieve satisfactory remission not only by working rapidly but also by providing a sustained response.


Subject(s)
Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Ketamine/pharmacology , Ketamine/therapeutic use , Signal Transduction/drug effects , Animals , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Humans , Models, Neurological , Time Factors
3.
Mol Psychiatry ; 17(8): 770-80, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21727899

ABSTRACT

The BH3-interacting domain death agonist (Bid) is a pro-apoptotic member of the B-cell lymphoma-2 (Bcl-2) protein family. Previous studies have shown that stress reduces levels of Bcl-2 in brain regions implicated in the pathophysiology of mood disorders, whereas antidepressants and mood stabilizers increase Bcl-2 levels. The Bcl-2 protein family has an essential role in cellular resilience as well as synaptic and neuronal plasticity and may influence mood and affective behaviors. This study inhibited Bid in mice using two pharmacological antagonists (BI-11A7 and BI-2A7); the selective serotonin reuptake inhibitor citalopram was used as a positive control. These agents were studied in several well-known rodent models of depression-the forced swim test (FST), the tail suspension test (TST), and the learned helplessness (LH) paradigm-as well as in the female urine sniffing test (FUST), a measure of sex-related reward-seeking behavior. Citalopram and BI-11A7 both significantly reduced immobility time in the FST and TST and attenuated escape latencies in mice that underwent the LH paradigm. In the FUST, both agents significantly improved duration of female urine sniffing in mice that had developed helplessness. LH induction increased the activation of apoptosis-inducing factor (AIF), a caspase-independent cell death constituent activated by Bid, and mitochondrial AIF expression was attenuated by chronic BI-11A7 infusion. Taken together, the results suggest that functional perturbation of apoptotic proteins such as Bid and, alternatively, enhancement of Bcl-2 function, is a putative strategy for developing novel therapeutics for mood disorders.


Subject(s)
Aniline Compounds/therapeutic use , Antidepressive Agents/therapeutic use , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Citalopram/therapeutic use , Depression/drug therapy , Drug Delivery Systems/psychology , Sulfonamides/therapeutic use , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacology , Apoptosis Inducing Factor/metabolism , Apoptosis Regulatory Proteins , Behavior, Animal/drug effects , Carrier Proteins/metabolism , Citalopram/administration & dosage , Cytochromes c/metabolism , Disease Models, Animal , Drug Delivery Systems/methods , Infusions, Intraventricular , Male , Mice , Mice, Inbred Strains , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Sulfides/administration & dosage , Sulfides/pharmacology , Sulfides/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
4.
Mol Psychiatry ; 15(12): 1152-63, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20308988

ABSTRACT

The subgranular zone of the adult hippocampal dentate gyrus contains a pool of neural stem cells that continuously divide and differentiate into functional granule cells. It has been shown that production of new hippocampal neurons is necessary for amelioration of stress-induced behavioral changes by antidepressants in animal models of depression. The survival of newly born hippocampal neurons is decreased by chronic psychosocial stress and increased by exposure to enriched environments. These observations suggest the existence of a link between hippocampal neurogenesis, stress-induced behavioral changes, and the beneficial effects of enriched environment. To show causality, we subjected transgenic mice with conditionally suppressed neurogenesis to psychosocial stress followed by environmental enrichment. First, we showed that repeated social defeat coupled with chronic exposure to an aggressor produces robust and quantifiable indices of submissive and depressive-like behaviors; second, subsequent exposure to an enriched environment led to extinction of the submissive phenotype, while animals exposed to an impoverished environment retained the submissive phenotype; and third, enrichment was not effective in reversing the submissive and depressive-like behaviors in transgenic mice lacking neurogenesis. Our data show two main findings. First, living in an enriched environment is highly effective in extinguishing submissive behavioral traits developed during chronic social stress, and second, these effects are critically dependent on adult neurogenesis, indicating that beneficial behavioral adaptations are dependent on intact adult neurogenesis.


Subject(s)
Adaptation, Physiological , Adult Stem Cells/cytology , Environment , Neural Stem Cells/cytology , Neurogenesis/physiology , Stress, Psychological/psychology , Adaptation, Psychological , Adult Stem Cells/physiology , Analysis of Variance , Animals , Cell Differentiation , Cell Tracking/methods , Dentate Gyrus/cytology , Depression/physiopathology , Depression/psychology , Disease Models, Animal , Dominance-Subordination , Female , Housing, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/physiology , Resilience, Psychological , Stress, Psychological/physiopathology
5.
Mol Psychiatry ; 15(9): 883-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20142820

ABSTRACT

Bipolar disorder (BPD) is characterized by vulnerability to episodic depression and mania and spontaneous cycling. Because of marked advances in candidate-gene and genome-wide association studies, the list of risk genes for BPD is growing rapidly, creating an unprecedented opportunity to understand the pathophysiology of BPD and to develop novel therapeutics for its treatment. However, genetic findings are associated with major unresolved issues, including whether and how risk variance leads to behavioral abnormalities. Although animal studies are key to resolving these issues, consensus is needed regarding how to define and monitor phenotypes related to mania, depression and mood swing vulnerability in genetically manipulated rodents. In this study we discuss multiple facets of this challenging area, including theoretical considerations, available tests, limitations associated with rodent behavioral modeling and promising molecular-behavioral findings. These include CLOCK, glycogen synthase kinase 3beta (GSK-3beta), glutamate receptor 6 (GluR6), extracellular signal-regulated kinase-1 (ERK1), p11 (or S100A10), vesicular monoamine transporter 2 (VMAT2 or SLC18A2), glucocorticoid receptors (GRs), Bcl-2-associated athanogene-1 (BAG1) and mitochondrial DNA polymerase-gamma (POLG). Some mutant rodent strains show behavioral clusters or activity patterns that cross-species phenocopy objective/observable facets of mood syndromes, and changes in these clustered behaviors can be used as outcome measures in genetic-behavioral research in BPD.


Subject(s)
Biomedical Research/trends , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Disease Models, Animal , Models, Genetic , Animals , Humans
6.
Mol Psychiatry ; 14(4): 448-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18227838

ABSTRACT

The extracellular signal-regulated kinase (ERK) pathway mediates neuronal plasticity in the CNS. The mood stabilizers lithium and valproate activate the ERK pathway in prefrontal cortex and hippocampus and potentiate ERK pathway-mediated neurite growth, neuronal survival and hippocampal neurogenesis. Here, we examined the role of the ERK pathway in behavioral plasticity related to facets of bipolar disorder. Mice with ERK1 ablation acquired reduced phosphorylation of RSK1, an ERK substrate, in prefrontal cortex and striatum, but not in hippocampus or cerebellum, indicating the ablation-induced brain region-specific ERK signaling deficits. ERK1 ablation produced a behavioral excitement profile similar to that induced by psychostimulants. The profile is characterized by hyperactivity, enhanced goal-directed activity and increased pleasure-related activity with potential harmful consequence. ERK1-ablated mice were hyperactive in multiple tests and resistant to behavioral despair in the forced swim test. These mice displayed more home-cage voluntary wheel running activities, rearings in a large arena and open-arm visits in an elevated plus maze. Treatments with valproate and olanzapine, but not lithium reduced baseline activities in ERK1-ablated mice. All three treatments attenuated amphetamine-induced hyperactivity in ablated mice. These data indicate a profound involvement of ERK1 signaling in behavioral excitement and in the behavioral action of antimanic agents. The extent to which ERK pathway perturbation contributes to the susceptibility, mood switch mechanism(s) and symptom pathophysiology of bipolar disorder requires further investigation. Whether there is a shared mechanism through which mood stabilizers produce their clinical actions on mood, thought and behavioral symptoms of mania also requires further investigation.


Subject(s)
Behavior, Animal/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Signal Transduction/physiology , Adjuvants, Immunologic , Administration, Oral , Amphetamine/pharmacology , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Central Nervous System Stimulants/pharmacology , Enzyme Inhibitors/pharmacology , Lithium Chloride/administration & dosage , Locomotion/drug effects , Locomotion/genetics , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/deficiency , Olanzapine , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/genetics , Swimming , Valproic Acid/pharmacology
7.
Mol Psychiatry ; 13(9): 858-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18332879

ABSTRACT

The glutamate receptor 6 (GluR6 or GRIK2, one of the kainate receptors) gene resides in a genetic linkage region (6q21) associated with bipolar disorder (BPD), but its function in affective regulation is unknown. Compared with wild-type (WT) and GluR5 knockout (KO) mice, GluR6 KO mice were more active in multiple tests and super responsive to amphetamine. In a battery of specific tests, GluR6 KO mice also exhibited less anxious or more risk-taking type behavior and less despair-type manifestations, and they also had more aggressive displays. Chronic treatment with lithium, a classic antimanic mood stabilizer, reduced hyperactivity, aggressive displays and some risk-taking type behavior in GluR6 KO mice. Hippocampal and prefrontal cortical membrane levels of GluR5 and KA-2 receptors were decreased in GluR6 KO mice, and chronic lithium treatment did not affect these decreases. The membrane levels of other glutamatergic receptors were not significantly altered by GluR6 ablation or chronic lithium treatment. Together, these biochemical and behavioral results suggest a unique role for GluR6 in controlling abnormalities related to the behavioral symptoms of mania, such as hyperactivity or psychomotor agitation, aggressiveness, driven or increased goal-directed pursuits, risk taking and supersensitivity to psychostimulants. Whether GluR6 perturbation is involved in the mood elevation or thought disturbance of mania and the cyclicity of BPD are unknown. The molecular mechanism underlying the behavioral effects of lithium in GluR6 KO mice remains to be elucidated.


Subject(s)
Bipolar Disorder/metabolism , Receptors, Kainic Acid/metabolism , Analysis of Variance , Animals , Antimanic Agents/therapeutic use , Avoidance Learning/drug effects , Behavioral Symptoms , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Exploratory Behavior/drug effects , Interpersonal Relations , Lithium Carbonate/therapeutic use , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Kainic Acid/deficiency , Risk-Taking , Swimming , Time Factors , GluK2 Kainate Receptor
9.
Science ; 306(5697): 882-4, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15514161

ABSTRACT

The prefrontal cortex is a higher brain region that regulates thought, behavior, and emotion using representational knowledge, operations often referred to as working memory. We tested the influence of protein kinase C (PKC) intracellular signaling on prefrontal cortical cognitive function and showed that high levels of PKC activity in prefrontal cortex, as seen for example during stress exposure, markedly impair behavioral and electrophysiological measures of working memory. These data suggest that excessive PKC activation can disrupt prefrontal cortical regulation of behavior and thought, possibly contributing to signs of prefrontal cortical dysfunction such as distractibility, impaired judgment, impulsivity, and thought disorder.


Subject(s)
Memory/physiology , Prefrontal Cortex/physiology , Protein Kinase C/metabolism , Adrenergic alpha-Agonists/pharmacology , Alkaloids , Animals , Benzophenanthridines , Carbolines/pharmacology , Electrophysiology , Enzyme Activation , Female , Imidazoles/pharmacology , Lithium Carbonate/pharmacology , Macaca mulatta , Male , Memory/drug effects , Neurons/drug effects , Neurons/physiology , Phenanthridines/pharmacology , Prefrontal Cortex/enzymology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/physiology , Signal Transduction , Stress, Physiological/physiopathology , Tetradecanoylphorbol Acetate/pharmacology , Valproic Acid/pharmacology
10.
Mol Psychiatry ; 9(8): 734-55, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15136794

ABSTRACT

Bipolar disorder afflicts approximately 1-3% of both men and women, and is coincident with major economic, societal, medical, and interpersonal consequences. Current mediations used for its treatment are associated with variable rates of efficacy and often intolerable side effects. While preclinical and clinical knowledge in the neurosciences has expanded at a tremendous rate, recent years have seen no major breakthroughs in the development of novel types of treatment for bipolar disorder. We review here approaches to develop novel treatments specifically for bipolar disorder. Deliberate (ie not by serendipity) treatments may come from one of two general mechanisms: (1) Understanding the mechanism of action of current medications and thereafter designing novel drugs that mimics these mechanism(s); (2) Basing medication development upon the hypothetical or proven underlying pathophysiology of bipolar disorder. In this review, we focus upon the first approach. Molecular and cellular targets of current mood stabilizers include lithium inhibitable enzymes where lithium competes for a magnesium binding site (inositol monophosphatase, inositol polyphosphate 1-phosphatase, glycogen synthase kinase-3 (GSK-3), fructose 1,6-bisphosphatase, bisphosphate nucleotidase, phosphoglucomutase), valproate inhibitable enzymes (succinate semialdehyde dehydrogenase, succinate semialdehyde reductase, histone deacetylase), targets of carbamazepine (sodium channels, adenosine receptors, adenylate cyclase), and signaling pathways regulated by multiple drugs of different classes (phosphoinositol/protein kinase C, cyclic AMP, arachidonic acid, neurotrophic pathways). While the task of developing novel medications for bipolar disorder is truly daunting, we are hopeful that understanding the mechanism of action of current mood stabilizers will ultimately lead clinical trials with more specific medications and thus better treatments those who suffer from this devastating illness.


Subject(s)
Antimanic Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Enzymes/genetics , Humans , Models, Biological
11.
Mol Psychiatry ; 9(8): 756-76, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15136795

ABSTRACT

Bipolar affective disorder (manic-depressive illness) is a common, severe, chronic, and often life-threatening illness, associated with significant comorbidity. The recognition of the significant morbidity and mortality of patients with bipolar disorder, as well as the growing appreciation that a high percentage of patients respond poorly to existing treatments, has made the task of discovering new therapeutic agents, that are both efficacious and have few side effects increasingly more important. Most recent agents introduced into the pharmacopeia for the treatment of bipolar disorder have been anticonvulsants and atypical antipsychotics. We propose that novel treatments developed specifically for bipolar disorder will arise from (1) understanding more precisely the molecular mechanisms of treatments that are clearly efficacious or (2) developing medications based on the knowledge obtained of the underlying pathophysiology of bipolar disorder. Knowledge with regard to the underlying pathophysiology of bipolar disorder is increasing at a rapid pace, including alterations in intracellular signaling cascades as well as impairments of cellular plasticity and resilience in critical neuronal circuits. We propose that therapeutics designed to enhance cellular plasticity and resilience and that counter maladaptive stress-responsive systems may have considerable utility for the treatment of bipolar disorder. Therapeutic strategies designed to address cellular resilience and plasticity include the regulation of neurotrophic pathways, glucocorticoid signaling, phosphodiesterase activity, and glutamatergic throughput and mitochondrial function. While the task of developing novel medications for bipolar disorder is truly daunting, these and similar approaches will ultimately lead to better medications for the millions who suffer from this devastating illness.


Subject(s)
Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Antimanic Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Bipolar Disorder/physiopathology , Brain/physiopathology , Humans , Mood Disorders/drug therapy , Mood Disorders/genetics , Mood Disorders/physiopathology , Neuronal Plasticity , Stress, Psychological
12.
Drug News Perspect ; 16(7): 453-63, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14668941

ABSTRACT

Despite the devastating impact that bipolar disorder has on the lives of millions worldwide, little is known for certain about its etiology or pathophysiology. Whereas research has traditionally focused on biogenic amines, it is becoming increasingly more apparent that intracellular pathways are involved in the etiology and treatment of the disease and that a true understanding of the pathophysiology of bipolar disorder must address its neurobiology at different physiological levels, that is, molecular, cellular, systems and behavioral levels. There is now considerable biochemical evidence that the antimanic agents lithium and valproate robustly activate the ERK signaling cascade in therapeutically relevant paradigms. This raises the possibility that this pathway may play a role in the antimanic effects of these agents. The present paper reviews behavioral studies that may shed light on the involvement of the ERK pathway in affective-like behaviors in animals. The available literature suggests that genetic manipulations of the brain-derived neurotrophic factor (BDNF)-ERK kinase pathway produces a variety of changes in affective-like behaviors, with most changes consistent with manic-like behavior. Thus, overall, mice with targeted mutation of the BDNF gene exhibited increased spontaneous locomotion and increased response to acute amphetamine, altered response to chronic cocaine, increased aggression, increase in risk-taking behavior, as demonstrated by time spent in the center of an open field, and changes in eating patterns. Although it has to be acknowledged that the currently available behavioral data from the BDNF-ERK pathway mutants is less than ideal to offer real substantiation relating this pathway to bipolar disorder, the data still supports the possibility that this pathway modulates manic-like behavior in animals, and perhaps mania in humans.


Subject(s)
Behavior, Animal/physiology , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Mitogen-Activated Protein Kinases/physiology , Signal Transduction/physiology , Animals , Behavior, Animal/drug effects , Bipolar Disorder/drug therapy , Humans , Lithium/therapeutic use , Mice , Mice, Mutant Strains , Signal Transduction/drug effects , Signal Transduction/genetics , Valproic Acid/therapeutic use
14.
Mol Psychiatry ; 7 Suppl 1: S46-56, 2002.
Article in English | MEDLINE | ID: mdl-11986995

ABSTRACT

The complexity of the unique biology of bipolar disorder--which includes the predisposition to episodic, and often progressive, mood disturbance--and the dynamic nature of compensatory processes in the brain, coupled with limitations in experimental design, have hindered our ability to identify the underlying pathophysiology of this fascinating neuropsychiatric disorder. Although we have yet to identify the specific abnormal genes in mood disorders, recent studies have implicated critical signal transduction pathways as being integral to the pathophysiology and treatment of bipolar disorder. In particular, a converging body of preclinical data has shown that chronic lithium and valproate, at therapeutically relevant concentrations, regulate the protein kinase C signaling cascade. This has led to the investigation of the antimanic efficacy of tamoxifen (at doses sufficient to inhibit protein kinase C), with very encouraging preliminary results. A growing body of data also suggests that impairments of neuroplasticity and cellular resilience may also underlie the pathophysiology of bipolar disorder. It is thus noteworthy that mood stabilizers, such as lithium and valproate, indirectly regulate a number of factors involved in cell survival pathways--including cAMP response element binding protein, brain derived neurotrophic factor, bcl-2 and mitogen-activated protein kinases--and may thus bring about some of their delayed long-term beneficial effects via under-appreciated neurotrophic effects. The development of novel treatments, which more directly target molecules involved in critical central nervous system cell survival and cell death pathways, has the potential to enhance neuroplasticity and cellular resilience, thereby modulating the long-term course and trajectory of these devastating illnesses.


Subject(s)
Anticonvulsants/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Mood Disorders/drug therapy , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/drug effects , Mood Disorders/metabolism , Neuronal Plasticity/physiology , Protein Kinase C/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects
15.
Biol Psychiatry ; 50(3): 217-24, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11513821

ABSTRACT

BACKGROUND: Studies suggest that lithium may have profound immunomodulatory effects in animal models as well as in humans. METHODS: In this study, whole blood cultures from normal control subjects were established for 5 days and the effects of lithium on cytokine production were investigated. Because many of lithium's actions have been postulated to be modulated through phosphoinositide (PI), protein kinase C (PKC) and cyclic adenosine monophosphate (c-AMP) signaling pathways, the effects of myo-inositol and prostaglandin E(2), alone or in combination with lithium, were also investigated. RESULTS: We found that lithium caused an increase in interleukin-4 and interleukin-10 levels, traditionally classified as T-helper lymphocyte type-2 cytokines, and a decrease in interleukin-2 and interferon-gamma levels, traditionally classified as T-helper lymphocyte type-1 (TH-1) cytokines. This shift cannot be fully explained by lithium's actions on the PI, PKC, or c-AMP messenger systems. CONCLUSIONS: Monocytes exposed to lithium in the presence of a mitogen for 5 days produced a shift toward the production of TH-2 cytokines and away from the production of TH-1 cytokines. The study suggests that lithium may have complex time-dependent effects on immune function.


Subject(s)
Antipsychotic Agents/pharmacology , Bipolar Disorder , Cytokines/metabolism , Lithium/pharmacology , Adult , Aged , Bipolar Disorder/drug therapy , Bipolar Disorder/immunology , Bipolar Disorder/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cytokines/immunology , Female , Humans , Male , Middle Aged , Phosphoric Monoester Hydrolases/metabolism
16.
Br J Psychiatry Suppl ; 41: s107-19, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11450170

ABSTRACT

BACKGROUND: New research is dramatically altering our understanding of the molecular mechanisms underlying neuronal communication. AIM: To elucidate the molecular mechanisms underlying the therapeutic effects of mood stabilizers. METHOD: Results from integrated clinical and laboratory studies are reviewed. RESULTS: Chronic administration of lithium and valproate produced a striking reduction in protein kinase C (PKC) isozymes in rat frontal cortex and hippocampus. In a small study, tamoxifen (also a PKC inhibitor) had marked antimanic efficacy. Both lithium and valproate regulate the DNA binding activity of the activator protein I family of transcription factors. Using mRNA differential display, it was also shown that chronic administration of lithium and valproate modulates expression of several genes. An exciting finding is that of a robust elevation in the levels of the cytoprotective protein, bcl-2. CONCLUSIONS: The results suggest that regulation of signalling pathways may play a major part in the long-term actions of mood stabilizers. Additionally, mood stabilizers may exert underappreciated neuroprotective effects.


Subject(s)
Antimanic Agents/pharmacology , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3 , Humans , Protein Kinase C/physiology , Signal Transduction/drug effects , Transcription Factors/drug effects
17.
Br J Psychiatry ; 178(Suppl 41): S107-19, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11388949

ABSTRACT

Background New research is dramatically altering our understanding of the molecular mechanisms underlying neuronal communication. Aim To elucidate the molecular mechanisms underlying the therapeutic effects of mood stabilisers. Method Results from integrated clinical and laboratory studies are reviewed. Results Chronic administration of lithium and valproate produced a striking reduction in protein kinase C (PKC) isozymes in rat frontal cortex and hippocampus. In a small study, tamoxifen (also a PKC inhibitor) had marked antimanic efficacy. Both lithium and valproate regulate the DNA binding activity of the activator protein 1 family of transcription factors. Using mRNA differential display, it was also shown that chronic administration of lithium and valproate modulates expression of several genes. An exciting finding is that of a robust elevation in the levels of the cytoprotective protein, bcl-2. Conclusions The results suggest that regulation of signalling pathways may play a major part in the long-term actions of mood stabilisers. Additionally, mood stabilisers may exert underappreciated neuroprotective effects.

18.
J Biol Chem ; 276(34): 31674-83, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11418608

ABSTRACT

The mood-stabilizing agents lithium and valproic acid (VPA) increase DNA binding activity and transactivation activity of AP-1 transcription factors, as well as the expression of genes regulated by AP-1, in cultured cells and brain regions involved in mood regulation. In the present study, we found that VPA activated extracellular signal-regulated kinase (ERK), a kinase known to regulate AP-1 function and utilized by neurotrophins to mediate their diverse effects, including neuronal differentiation, neuronal survival, long term neuroplasticity, and potentially learning and memory. VPA-induced activation of ERK was blocked by the mitogen-activated protein kinase/ERK kinase inhibitor PD098059 and dominant-negative Ras and Raf mutants but not by dominant-negative stress-activated protein kinase/ERK kinase and mitogen-activated protein kinase kinase 6 mutants. VPA also increased the expression of genes regulated by the ERK pathway, including growth cone-associated protein 43 and Bcl-2, promoted neurite growth and cell survival, and enhanced norepinephrine uptake and release. These data demonstrate that VPA is an ERK pathway activator and produces neurotrophic effects.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neurites/drug effects , Valproic Acid/pharmacology , Base Sequence , Cell Survival , Culture Media, Serum-Free , DNA Primers , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , GAP-43 Protein/metabolism , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Norepinephrine/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein
19.
Nat Med ; 7(5): 541-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11329053

ABSTRACT

Major depressive disorders, long considered to be of neurochemical origin, have recently been associated with impairments in signaling pathways that regulate neuroplasticity and cell survival. Agents designed to directly target molecules in these pathways may hold promise as new therapeutics for depression.


Subject(s)
Depressive Disorder/pathology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cell Survival/drug effects , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Glucocorticoids/pharmacology , Neuronal Plasticity/drug effects , Signal Transduction
20.
Psychopharmacol Bull ; 35(2): 5-49, 2001.
Article in English | MEDLINE | ID: mdl-12397885

ABSTRACT

Mood disorders have traditionally been conceptualized as neurochemical disorders, but there is now evidence from a variety of sources demonstrating regional reductions in central nervous system (CNS) volume, as well as reductions in the numbers and/or sizes of glia and neurons in discrete brain areas. Although the precise cellular mechanisms underlying these morphometric changes remain to be fully elucidated, the data suggests that mood disorders are associated with impairments of structural plasticity and cellular resilience. Recent preclinical and clinical studies have shown that signaling pathways involved in regulating cell survival and cell death are long-term targets for the actions of antidepressants and mood stabilizers. Antidepressants, lithium, and valproate indirectly regulate a number of factors involved in cell survival pathways, including CREB, BDNF, Bcl-2, and MAP kinases, and may thus bring about some of their delayed long term beneficial effects via underappreciated neurotrophic effects. The future development of treatments that more directly target molecules involved in critical CNS cell survival and cell death pathways thus hold promise as novel, improved long-term treatments for mood disorders.


Subject(s)
Mood Disorders/drug therapy , Mood Disorders/pathology , Neuronal Plasticity/physiology , Psychotropic Drugs/pharmacology , Animals , Antidepressive Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Atrophy , Bipolar Disorder/pathology , Brain/pathology , Cell Membrane/drug effects , Cell Membrane/pathology , Cell Membrane/physiology , Cell Survival/drug effects , Glycogen Synthase Kinases/physiology , Hippocampus/pathology , Humans , Lithium/pharmacology , Magnetic Resonance Imaging , Neuroprotective Agents/pharmacology , Psychotropic Drugs/therapeutic use , Signal Transduction/drug effects , Stress, Psychological/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...