Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
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.
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
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
8.
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
9.
Arch Gen Psychiatry ; 52(7): 531-43, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7598629

ABSTRACT

Lithium remains the most widely used treatment for bipolar disorder, and this monovalent cation represents one of psychiatry's most important treatments. Despite its demonstrated efficacy in reducing both the frequency and severity of recurrent affective episodes and decades of clinical use, the molecular mechanisms underlying its therapeutic actions have not fully been elucidated. In this report, we review the exciting recent progress in the identification of key components of signal transduction pathways (in particular, guanine nucleotide-binding proteins [G proteins], adenylyl cyclases, and protein kinase C isozymes) as targets for lithium's actions and attempt to integrate these effects with the large body of data emphasizing alterations in various neurotransmitter (particularly monoaminergic) systems. Regulation of signal transduction within critical regions of the brain by lithium affects the function of multiple neurotransmitter systems and may thus explain lithium's efficacy in protecting susceptible individuals from spontaneous, stress-induced, and drug-induced cyclic affective episodes. Recent evidence has also demonstrated significant effects of lithium on the regulation of gene expression in the central nervous system, effects that may play a major role in the long-term stabilization of mood. The identification of these intracellular targets for lithium's actions offers the potential for the development of novel, improved therapeutic agents and, in conjunction with molecular genetic approaches, may facilitate our understanding of the biological factors predisposing individuals to manic-depressive illness.


Subject(s)
Lithium/pharmacology , Signal Transduction/drug effects , Adenylyl Cyclases/drug effects , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Brain/drug effects , Brain/enzymology , Cyclic AMP/metabolism , GTP-Binding Proteins/drug effects , Gene Expression/drug effects , Humans , Isoenzymes/drug effects , Lithium/therapeutic use , Protein Kinase C/drug effects , Receptors, Adrenergic, beta/drug effects , Second Messenger Systems/drug effects
10.
Arch Gen Psychiatry ; 48(6): 505-12, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1645513

ABSTRACT

The effects of 2 weeks of lithium carbonate administration at therapeutic plasma levels were examined in 11 normal volunteers. Serotoninergic function before and after lithium administration was assessed using low-dose intravenous clomipramine hydrochloride challenge, while urinary and plasma metabolites of norepinephrine (NE) were used to assess noradrenergic systems. Long-term lithium administration in normal subjects did not significantly or consistently enhance serotonin-mediated neuroendocrine responses but did increase measures related to neuronal release of NE. No statistically significant effects of lithium on prolactin, corticotropin, or cortisol responses to serotoninergic challenge could be detected. The probability of a type II error was assessed, and a doubling of prolactin level was unlikely to have been missed, although more modest increases (less than 75%) could have been overlooked. After 2 weeks of lithium administration, there were significant increases in 24-hour urinary excretion of NE, normetanephrine, and fractional NE release, compatible with increased neuronal release of NE and a lithium-induced subsensitivity in alpha 2-adrenergic receptor function. These changes were not statistically significant after 1 week of administration, suggesting that increased NE release is characteristic of long- rather than short-term lithium administration. Since previous reports have demonstrated enhanced prolactin responses after short- but not long-term lithium use, the present study points to temporal specificity in lithium's effects on both serotoninergic and noradrenergic function. Lithium's effects on NE release were consistent but small (a 16% increase), while its effects on serotoninergic responses were larger (a 50% increase in prolactin responses) but quite inconsistent, suggesting that neither of these systems is the primary site of action of lithium.


Subject(s)
Lithium/pharmacology , Norepinephrine/metabolism , Serotonin/physiology , Adrenocorticotropic Hormone/blood , Adult , Blood Pressure/drug effects , Clomipramine/administration & dosage , Clomipramine/pharmacology , Female , Heart Rate/drug effects , Humans , Hydrocortisone/blood , Infusions, Intravenous , Lithium Carbonate , Male , Methoxyhydroxyphenylglycol/urine , Norepinephrine/urine , Normetanephrine/urine , Prolactin/blood , Receptors, Adrenergic/drug effects , Receptors, Serotonin/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiology , Vanilmandelic Acid/urine
11.
Arch Gen Psychiatry ; 52(2): 135-44, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7848049

ABSTRACT

BACKGROUND: This study examines recent suggestions from a number of investigators that signal-transducing guanine nucleotide-binding (G) proteins may be involved in the pathophysiology of bipolar affective disorder and may represent molecular targets for lithium's mood-stabilizing actions. METHODS: We used selective antibodies to quantitate the levels of the G protein alpha subunits that regulate adenylate cyclase activity (G alpha s and G alpha i2) and phosphoinositide turnover (G alpha q/11). We also quantitated levels of pertussis toxin-catalyzed phosphate 32-labeled adenosine diphosphate ([32P]ADP) ribosylation in platelet and leukocyte membranes from a group of 14 untreated (predominantly manic) patients with bipolar affective disorder, 20 lithium-treated euthymic patients with bipolar affective disorder, and 11 healthy controls. RESULTS: In both tissues, the immunolabeling of the 45-kd form of G alpha s was higher in the bipolar affective disorder group considered as a whole (treated or untreated) compared with controls, effects that reached statistical significance in the leukocyte membranes. There were no significant differences in the immunolabeling of G alpha i1/2, G alpha q/11, or pertussis toxin-catalyzed [32P]ADP ribosylation in either tissue in the untreated bipolar affective disorder group compared with controls. In both tissues, lithium-treated subjects demonstrated lower levels of G alpha q/11 and higher levels of pertussis toxin-catalyzed [32P]ADP-ribosylation, which reached significance in the platelet membranes. CONCLUSIONS: Our results are complementary to the previously reported findings of elevated G alpha s levels in postmortem brain tissue form patients with bipolar affective disorder and in mononuclear leukocytes obtained from depressed patients with bipolar (but not unipolar) affective disorder. The significantly higher levels of pertussis toxin-catalyzed [32P]ADP ribosylation in the subjects receiving long-term lithium-treatment replicates our findings in rat cortex and in healthy volunteers and adds to the growing body of evidence implicating G alpha i as a target of lithium's actions.


Subject(s)
Bipolar Disorder/physiopathology , GTP-Binding Proteins/physiology , Adenosine Diphosphate Ribose/metabolism , Adenylate Cyclase Toxin , Adult , Bipolar Disorder/blood , Bipolar Disorder/drug therapy , Blood Platelets/chemistry , Blood Platelets/metabolism , Female , Humans , Immunoblotting , Leukocytes/chemistry , Leukocytes/metabolism , Lithium/pharmacology , Lithium/therapeutic use , Male , Middle Aged , Pertussis Toxin , Phosphorus Radioisotopes/metabolism , Virulence Factors, Bordetella/metabolism
12.
Arch Gen Psychiatry ; 48(6): 513-24, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1645514

ABSTRACT

As part of a study of the effects of lithium carbonate on neurochemical function in man, platelet and lymphocyte adenylate cyclase activity and lymphocyte beta-adrenergic receptor binding characteristics were determined before and after 2 weeks of lithium treatment in 10 normal volunteers. Lithium had differential effects on platelet and lymphocyte adenylate cyclase activity. In platelets, basal and stimulated (guanyl imidodiphosphate [Gpp[NH]p] or cesium fluoride) adenylate cyclase activity was significantly augmented by lithium treatment. By contrast, in lymphocytes, Gpp(NH)p- and cesium fluoride-stimulated adenylate cyclase activity was unaffected, while basal activity was decreased modestly after lithium. These results are consistent with preclinical studies that suggest that lithium's effects on adenylate cyclase activity are specific with respect to tissue and brain region and that lithium may interfere with guanine nucleotide binding (G) protein function. Lithium treatment significantly increased the ratio of low- to high-affinity dissociation constants for agonist displacement of antagonist binding to lymphocyte beta-adrenergic receptors (thought to reflect coupling between the beta-adrenergic receptor and stimulatory G protein). Lithium had significant effects on measures associated with signal transduction that might be contrasted to its more subtle effects on neuronal function (norepinephrine release) and neuroendocrine systems (responses to serotoninergic challenge) in these same subjects (reported in a companion article). Lithium's primary site of action may be on signal transduction mechanisms. These effects subsequently may be manifested in changes in neurotransmitter function that may be important to lithium's mood-stabilizing actions.


Subject(s)
Adenylyl Cyclases/metabolism , Fluorides , Lithium/pharmacology , Receptors, Adrenergic, beta/metabolism , Adult , Affect/drug effects , Blood Platelets/drug effects , Blood Platelets/enzymology , Cesium/metabolism , Female , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Guanylyl Imidodiphosphate/metabolism , Humans , Lithium/blood , Lithium/metabolism , Lithium Carbonate , Lymphocytes/drug effects , Lymphocytes/enzymology , Male , Nerve Tissue Proteins/metabolism , Norepinephrine/metabolism , Prospective Studies , Receptors, Adrenergic, beta/drug effects , Second Messenger Systems/drug effects , Serotonin/pharmacology , Serotonin/physiology , Signal Transduction/drug effects
13.
Arch Gen Psychiatry ; 54(1): 44-8, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9006399

ABSTRACT

BACKGROUND: The available evidence for an involvement of the heterotrimeric guanine-nucleotide-binding proteins (G proteins) in bipolar disorder relies primarily on the effects of lithium salts on G protein function and on alterations in the concentration or function of G proteins (most notably Gs-alpha) in peripheral leukocytes and in postmortem tissues of patients with bipolar disorder. METHODS: The hypothesis that a mutation in Gs-alpha gene confers an increased susceptibility to bipolar disorder was tested by the following strategies: (1) mutational screening of the Gs-alpha subunit gene coding sequences and promoter sequences by denaturing gradient gel electrophoresis in unrelated individuals with bipolar disorder and (2) association and linkage analyses with a common silent exonic polymorphism, using genetic allelic information from American families with at least 1 affected child. For association analysis, the transmission test for linkage disequilibrium was used; for linkage analysis, nonparametric methods were used. RESULTS: No structural or regulatory mutations in this gene were found in bipolar disorder; the results of association and genetic linkage were negative. CONCLUSION: Our results do not support the speculation that the Gs-alpha protein gene has a role in the genetic predisposition to bipolar disorder.


Subject(s)
Bipolar Disorder/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation , Base Sequence , DNA Mutational Analysis , Exons , Genetic Linkage , Genetic Predisposition to Disease , Humans , Linkage Disequilibrium , Lod Score , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Genetic , Psychotic Disorders/genetics , Risk Factors
14.
Biol Psychiatry ; 32(7): 549-79, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1333286

ABSTRACT

Signal-transducing G proteins, heterotrimers formed of alpha, beta, and gamma subunits, are central to the coordination of receptor-effector communication. They are derived from a large gene family, and recent cloning and sequencing of cDNAs encoding the alpha subunits, which confer receptor and effector specificity on the heterotrimer, have defined four major classes, Gs, Gi, Gq, and G12, with at least 16 isotypes. The G proteins that coordinate receptor-effector activity are especially important in the central nervous system (CNS), where they serve widespread, critical roles in the regulation of neuronal function, maintain the functional balance between neurotransmitter systems, and, as such, represent attractive potential targets for antidepressant drugs. We describe an integrated series of animal and cell culture studies aimed at testing the hypothesis that alterations in G protein function may contribute the complex neuroadaptive mechanisms involved in the clinical actions of antidepressants, and demonstrate that long-term administration of a wide spectrum of antidepressant drugs regulate G alpha s, G alpha i1, G alpha i2, G alpha o, G alpha q, and G alpha 12 mRNA and protein expression in various areas of the rat brain. Additionally, we present the polymerase chain reaction-(PCR) mediated cross-species partial cDNA cloning and sequencing of rat and human G alpha o and rat G alpha 12, illustrate the regional distribution of G alpha mRNA and protein in rat brain, and provide evidence that different classes of antidepressants alter expression and/or stability of the recently identified G alpha 12 mRNA. We conclude that long-term treatment with antidepressant drugs exerts differential effects on G alpha mRNA and protein expression in rat brain, thus modifying signal transduction as an integral part of complex neuroadaptive mechanisms that may underlie their therapeutic efficacy. The development of novel drugs with G proteins as primary targets remains an attractive prospect for the future.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , GTP-Binding Proteins/drug effects , Gene Expression Regulation/drug effects , Signal Transduction/drug effects , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Blotting, Northern , Brain/physiology , Cell Line , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , GTP-Binding Proteins/classification , GTP-Binding Proteins/genetics , Gene Expression Regulation/physiology , Glioma , Humans , Male , Molecular Sequence Data , RNA Probes , RNA, Messenger/drug effects , RNA, Messenger/genetics , Rats , Rats, Wistar , Signal Transduction/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
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.
Biol Psychiatry ; 48(6): 518-30, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11018224

ABSTRACT

Clinical studies over the years have provided evidence that monoamine signaling and hypothalamic-pituitary-adrenal axis disruption are integral to the pathophysiology of bipolar disorder. A full understanding of the pathophysiology from a molecular to a systems level must await the identification of the susceptibility and protective genes driving the underlying neurobiology of bipolar disorder. Furthermore, the complexity of the unique biology of this affective 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 progress to date. Imaging studies in patient populations have provided evidence of a role for anterior cingulate, amygdala, and prefrontal cortex in the pathophysiology of bipolar disorder. More recent research strategies designed to uncover the molecular mechanisms underlying our pharmacologic treatments and their interaction in the regulation of signal transduction as well as more advanced brain imaging studies remain promising approaches. This experimental strategy provides data derived from the physiologic response of the system in affected individuals and addresses the critical dynamic interaction with pharmacologic agents that effectively modify the clinical expression of the pathophysiology.


Subject(s)
Antipsychotic Agents/pharmacology , Bipolar Disorder/metabolism , Brain/drug effects , Brain/metabolism , Signal Transduction/drug effects , Bipolar Disorder/drug therapy , Bipolar Disorder/physiopathology , Brain/physiopathology , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/metabolism , Humans , Ion Pumps/metabolism , Lithium/pharmacology , Neural Pathways/drug effects , Neural Pathways/metabolism , Protein Kinase C/metabolism
17.
Biol Psychiatry ; 46(10): 1328-51, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10578449

ABSTRACT

Understanding the biology of the pharmacological stabilization of mood will undoubtedly serve to provide significant insight into the pathophysiology of manic-depressive illness (MDI). Accumulating evidence from our laboratories and those of other researchers has identified the family of protein kinase C isozymes as a shared target in the brain for the long-term action of both lithium and valproate. In rats chronically treated with lithium, there is a reduction in the hippocampus of the expression of two protein kinase isozymes, alpha and epsilon, as well as a reduction in the expression of a major PKC substrate, MARCKS, which has been implicated in long-term neuroplastic events in the developing and adult brain. In addition, we have been investigating the down-stream impact of these mood stabilizers on another kinase system, GSK-3 beta and on the AP-1 family of transcription factors. Further studies have generated promising preliminary data in support of the antimanic action of tamoxifen, and antiestrogen that is also a PKC inhibitor. Future studies must address the therapeutic relevance of these protein targets in the brain using innovative strategies in both animal and clinical investigations to ultimately create opportunities for the discovery of the next generations of mood stabilizers for the treatment of MDI.


Subject(s)
Awards and Prizes , Bipolar Disorder/drug therapy , Bipolar Disorder/enzymology , Brain/drug effects , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/therapeutic use , Enzyme Activators/therapeutic use , Protein Kinase C/metabolism , Research , Signal Transduction/drug effects , Animals , Antimanic Agents/pharmacology , Antimanic Agents/therapeutic use , Binding, Competitive , Bipolar Disorder/genetics , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/pharmacology , Disease Models, Animal , Enzyme Activators/pharmacology , Glycogen Synthase Kinase 3 , Humans , Lithium/pharmacology , Lithium/therapeutic use , Male , Protein Kinase C/genetics , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics , Transcription, Genetic/genetics , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
18.
Biol Psychiatry ; 46(7): 929-40, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10509176

ABSTRACT

Recent advances in cellular and molecular biology have resulted in the identification of two novel, hitherto completely unexpected targets of lithium's actions, discoveries that may have a major impact on the future use of this unique cation in biology and medicine. Chronic lithium treatment has been demonstrated to markedly increase the levels of the major neuroprotective protein, bcl-2 in rat frontal cortex, hippocampus, and striatum. Similar lithium-induced increases in bcl-2 are also observed in cells of human neuronal origin, and are observed in rat frontal cortex at lithium levels as low as approximately 0.3 mmol/L. Bcl-2 is widely regarded as a major neuroprotective protein, and genetic strategies that increase bcl-2 levels have demonstrated not only robust protection of neurons against diverse insults, but have also demonstrated an increase the regeneration of mammalian CNS axons. Lithium has also been demonstrated to inhibit glycogen synthase kinase 3 beta (GSK-3 beta), an enzyme known to regulate the levels of phosphorylated tau and beta-catenin (both of which may play a role in the neurodegeneration observed in Alzheimer's disease). Consistent with the increases in bcl-2 levels and inhibition of GSK-3 beta, lithium has been demonstrated to exert robust protective effects against diverse insults both in vitro and in vivo. These findings suggest that lithium may exert some of its long term beneficial effects in the treatment of mood disorders via underappreciated neuroprotective effects. To date, lithium remains the only medication demonstrated to markedly increase bcl-2 levels in several brain areas; in the absence of other adequate treatments, the potential efficacy of lithium in the long term treatment of certain neurodegenerative disorders may be warranted.


Subject(s)
Antimanic Agents/pharmacology , Lithium Carbonate/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Survival/drug effects , Gene Expression/genetics , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Nerve Regeneration/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Signal Transduction/drug effects
19.
Biol Psychiatry ; 48(8): 740-54, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11063971

ABSTRACT

Recent neuroimaging studies have demonstrated regional central nervous system volume reductions in mood disorders, findings that are complemented by postmortem observations of cell atrophy and loss. It is thus noteworthy that lithium and valproate have recently been demonstrated to robustly increase the expression of the cytoprotective protein bcl-2 in the central nervous system. Chronic lithium not only exerts neuroprotective effects in several preclinical paradigms but also enhances hippocampal neurogenesis. Valproate robustly promotes neurite outgrowth and activates the ERK mitogen-activated protein kinase pathway, a signaling pathway utilized by many endogenous neurotrophic factors. Consistent with its preclinical neurotrophic/neuroprotective effects, chronic lithium treatment of patients with manic-depressive illness increases brain N-acetylaspartate (a putative marker of neuronal viability and function) levels, an effect that is localized almost exclusively to gray matter. To determine if lithium was producing neuropil increases, quantitative three-dimensional magnetic resonance imaging studies were undertaken, which revealed that chronic lithium significantly increases total gray matter volume in the human brain of patients with manic-depressive illness. Together, these results suggest that a reconceptualization about the optimal long-term treatment of recurrent mood disorders is warranted. Optimal long-term treatment for these severe illnesses may only be achieved by the early use of agents with neurotrophic/neuroprotective effects, irrespective of the primary, symptomatic treatment.


Subject(s)
Affect/drug effects , Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/physiopathology , Lithium/therapeutic use , Neuroprotective Agents/therapeutic use , Valproic Acid/therapeutic use , Antimanic Agents/pharmacology , Atrophy/etiology , Atrophy/pathology , Bipolar Disorder/complications , Brain/pathology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death/drug effects , Cell Death/physiology , Genes, bcl-2/genetics , Humans , Lithium/pharmacology , Magnetic Resonance Imaging , Neuroprotective Agents/pharmacology , RNA, Messenger/genetics , Valproic Acid/pharmacology
20.
Biol Psychiatry ; 48(1): 1-8, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10913502

ABSTRACT

BACKGROUND: Recent preclinical studies have shown that lithium (Li) robustly increases the levels of the major neuroprotective protein, bcl-2, in rat brain and in cells of human neuronal origin. These effects are accompanied by striking neuroprotective effects in vitro and in the rodent central nervous system in vivo. We have undertaken the present study to determine if lithium exerts neurotrophic/ neuroprotective effects in the human brain in vivo. METHODS: Using quantitative proton magnetic resonance spectroscopy, N-acetyl-aspartate (NAA) levels (a putative marker of neuronal viability and function) were investigated longitudinally in 21 adult subjects (12 medication-free bipolar affective disorder patients and 9 healthy volunteers). Regional brain NAA levels were measured at baseline and following 4 weeks of lithium (administered in a blinded manner). RESULTS: A significant increase in total brain NAA concentration was documented (p < .0217). NAA concentration increased in all brain regions investigated, including the frontal, temporal, parietal, and occipital lobes. CONCLUSIONS: This study demonstrates for the first time that Li administration at therapeutic doses increases brain NAA concentration. These findings provide intriguing indirect support for the contention that chronic lithium increases neuronal viability/function in the human brain, and suggests that some of Li's long-term beneficial effects may be mediated by neurotrophic/neuroprotective events.


Subject(s)
Aspartic Acid/analogs & derivatives , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Brain/drug effects , Genes, bcl-2/drug effects , Lithium/pharmacology , Neuroprotective Agents/pharmacology , Adult , Analysis of Variance , Aspartic Acid/drug effects , Aspartic Acid/metabolism , Bipolar Disorder/genetics , Brain/metabolism , Double-Blind Method , Female , Gene Expression Regulation/drug effects , Humans , Lithium/therapeutic use , Magnetic Resonance Spectroscopy , Male , Middle Aged , Neuroprotective Agents/therapeutic use , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL