Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Neuroinflammation ; 19(1): 292, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36482444

ABSTRACT

The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.


Subject(s)
Neuronal Plasticity , Male , Animals , Mice
2.
Front Pharmacol ; 11: 603979, 2020.
Article in English | MEDLINE | ID: mdl-33613281

ABSTRACT

Vortioxetine is a novel multimodal antidepressant that modulates a wide range of neurotransmitters throughout the brain. Preclinical and clinical studies have shown that vortioxetine exerts positive effects on different cognitive domains and neuroprotective effects. Considering the key role of microglial cells in brain plasticity and cognition, we aimed at investigating the effects of pretreatment with vortioxetine in modulating behavioral and molecular effects induced by an immune challenge: peripheral injection of lipopolysaccharide (LPS). To this purpose, C57BL/6J male mice were first exposed to a 28-day standard diet or vortioxetine-enriched diet, which was followed by an acute immune challenge with LPS. Sickness symptoms and depressive-like behaviors (anhedonia and memory impairment) were tested 6 and 24 h after exposure to LPS, respectively. Moreover, the expressions of markers of immune activation and M1/M2 markers of microglia polarization were measured in the dorsal and ventral parts of the hippocampus. The pretreatment with vortioxetine did not affect both LPS-induced sickness behavior and anhedonia but prevented the deficit in the recognition memory induced by the immune challenge. At the transcriptional level, chronic exposure to vortioxetine did not prevent LPS-induced upregulation of proinflammatory cytokines 6 h after the immune challenge but rather seemed to potentiate the immune response to the challenge also by affecting the levels of expression of markers of microglia M1 phenotype, like cluster of differentiation (CD)14 and CD86, in an area-dependent manner. However, at the same time point, LPS injection significantly increased the expression of the M2 polarization inducer, interleukin 4, only in the hippocampus of animals chronically exposed to vortioxetine. These results demonstrate that a chronic administration of vortioxetine specifically prevents LPS-induced memory impairment, without affecting acute sickness behavior and anhedonia, and suggest that hippocampal microglia may represent a cellular target of this novel antidepressant medication. Moreover, we provide a useful model to further explore the molecular mechanisms specifically underlying cognitive impairments following an immune challenge.

3.
Neurosci Biobehav Rev ; 108: 602-616, 2020 01.
Article in English | MEDLINE | ID: mdl-31786320

ABSTRACT

The purpose of this review is to illustrate how a reductionistic, but sophisticated, approach based on the use of a simple model system such as the pond snail Lymnaea stagnalis (L. stagnalis), might be useful to address fundamental questions in learning and memory. L. stagnalis, as a model, provides an interesting platform to investigate the dialog between the synapse and the nucleus and vice versa during memory and learning. More importantly, the "molecular actors" of the memory dialogue are well-conserved both across phylogenetic groups and learning paradigms, involving single- or multi-trials, aversion or reward, operant or classical conditioning. At the same time, this model could help to study how, where and when the memory dialog is impaired in stressful conditions and during aging and neurodegeneration in humans and thus offers new insights and targets in order to develop innovative therapies and technology for the treatment of a range of neurological and neurodegenerative disorders.


Subject(s)
Aging , Lymnaea , Memory Disorders , Memory , Models, Animal , Neurosciences , Stress, Psychological , Translational Research, Biomedical , Aging/physiology , Animals , Memory/physiology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Stress, Psychological/metabolism
4.
Behav Brain Res ; 364: 140-148, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30771367

ABSTRACT

Understanding resilience is a major challenge to improve current pharmacological therapies aimed at complementing psychological-based approaches of stress-related disorders. In particular, resilience is a multi-factorial construct where the complex network of molecular events that drive the process still needs to be resolved. Here, we exploit the acute escape deficit model, an animal model based on exposure to acute unavoidable stress followed by an escape test, to define vulnerable and resilient phenotypes in rats. Hippocampus and prefrontal cortex (PFC), two of the brain areas most involved in the stress response, were analysed for gene expression at two different time points (3 and 24 h) after the escape test. Total Brain-Derived Neurotrophic Factor (BDNF) was highly responsive in the PFC at 24-h after the escape test, while expression of BDNF transcript IV increased in the hippocampus of resistant animals 3 h post-test. Expression of memory enhancers like Neuronal PAS Domain Protein 4 (Npas4) and Activity-regulated cytoskeleton-associated protein (Arc) decreased in a time- and region-dependent fashion in both behavioural phenotypes. Also, the memory inhibitor Protein Phosphatase 1 (Ppp1ca) was increased in the hippocampus of resilient rats at 3 h post-test. Given the importance of neurotrophic factors and synaptic plasticity-related genes for the development of appropriate coping strategies, our data contribute to an additional step forward in the comprehension of the psychobiology of stress and resiliency.


Subject(s)
Adaptation, Psychological/physiology , Brain-Derived Neurotrophic Factor/metabolism , Escape Reaction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cytoskeletal Proteins/metabolism , Gene Expression , Hippocampus/metabolism , Male , Models, Animal , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/metabolism
7.
Mol Psychiatry ; 22(4): 552-561, 2017 04.
Article in English | MEDLINE | ID: mdl-26645631

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) represent the most common treatment for major depression. However, their efficacy is variable and incomplete. In order to elucidate the cause of such incomplete efficacy, we explored the hypothesis positing that SSRIs may not affect mood per se but, by enhancing neural plasticity, render the individual more susceptible to the influence of the environment. Consequently, SSRI administration in a favorable environment promotes a reduction of symptoms, whereas in a stressful environment leads to a worse prognosis. To test such hypothesis, we exposed C57BL/6 mice to chronic stress in order to induce a depression-like phenotype and, subsequently, to fluoxetine treatment (21 days), while being exposed to either an enriched or a stressful condition. We measured the most commonly investigated molecular, cellular and behavioral endophenotypes of depression and SSRI outcome, including depression-like behavior, neurogenesis, brain-derived neurotrophic factor levels, hypothalamic-pituitary-adrenal axis activity and long-term potentiation. Results showed that, in line with our hypothesis, the endophenotypes investigated were affected by the treatment according to the quality of the living environment. In particular, mice treated with fluoxetine in an enriched condition overall improved their depression-like phenotype compared with controls, whereas those treated in a stressful condition showed a distinct worsening. Our findings suggest that the effects of SSRI on the depression- like phenotype is not determined by the drug per se but is induced by the drug and driven by the environment. These findings may be helpful to explain variable effects of SSRI found in clinical practice and to device strategies aimed at enhancing their efficacy by means of controlling environmental conditions.


Subject(s)
Fluoxetine/metabolism , Fluoxetine/pharmacology , Affect/drug effects , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Depression/drug therapy , Depressive Disorder, Major/drug therapy , Endophenotypes , Environment , Hypothalamo-Hypophyseal System/drug effects , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology
8.
J Neurosci Res ; 84(8): 1789-98, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17016858

ABSTRACT

The objective of this study was to test whether postnatal chronic inflammation resulted in altered reactivity to pain later in life when reexposed to the same inflammatory agent and whether this alteration correlated with brain-region-specific patterns of N-methyl-D-aspartate (NMDA) receptor subtype gene expression. Neonatal mouse pups received a single injection of complete Freund's adjuvant (CFA) or saline into the left hind paw on postnatal day 1 or 14. At 12 weeks of age, both neonatal CFA- and saline-treated animals received a unilateral injection of CFA in the left hind paw. Adult behavioral responsiveness of the left paw to a radiant heat source was determined in mice treated neonatally with saline or CFA before and after receiving CFA as adults. Twenty-four hours later, brains were dissected and NMDA receptor subunit gene expression was determined in four different brain areas by using an RNase protection assay. The results indicated that NMDA receptor subtype gene expression in adult mice exposed to persistent neonatal peripheral inflammation was brain region specific and that NMDA gene expression and pain reactivity differed according to the day of neonatal CFA exposure. Similarly, adult behavioral responsiveness to a noxious radiant heat source differed according to the age of neonatal exposure to CFA. The data suggest a possible molecular basis for the hypothesis that chronic persistent inflammation experienced early during development may permanently alter the future behavior and the sensitivity to pain later in life, especially in response to subsequent or recurrent inflammatory events.


Subject(s)
Central Nervous System , Gene Expression Regulation, Developmental/physiology , Inflammation/complications , Pain/etiology , Receptors, N-Methyl-D-Aspartate/metabolism , Age Factors , Animals , Animals, Newborn , Behavior, Animal , Central Nervous System/growth & development , Central Nervous System/metabolism , Central Nervous System/pathology , Female , Freund's Adjuvant , Gene Expression Regulation, Developmental/drug effects , Mice , Pain/metabolism , Pain Measurement , Protein Subunits/metabolism , Reaction Time/physiology , Ribonucleases/pharmacology , Time Factors
9.
J Affect Disord ; 79 Suppl 1: S15-20, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15121343

ABSTRACT

The mechanism underlying the therapeutic action of mood stabilizers in bipolar disorder is not completely understood. The discovery that anticonvulsant agents, such as valproate (VPA), were effective in the treatment of bipolar disorder suggested a common biochemical mechanism(s) with lithium. Recent research has focused on how VPA and lithium change the activities of cellular signal transduction systems, especially the cyclic AMP and phosphoinositide second messenger pathways. Despite being structurally dissimilar, VPA produces effects on the protein kinase C (PKC) signalling pathway that are similar to lithium, although the VPA effects appear to be largely independent of myo-inositol. Furthermore, the therapeutic benefit of either drug require a prolonged administration suggesting alterations at the genomic level. Studies have revealed that both VPA and lithium altered the expression of several early inducible genes belonging to the AP-1 family of transcription factors; this family is responsible for controlling the expression of a number of genes including cytoprotective proteins such as the anti-apoptotic protein, bcl-2. Evidence shows that chronic administration of VPA or lithium can stimulate bcl-2 expression as well as inhibit GSK-3 beta activity, which renders a cell less susceptible to apoptosis. Thus, the mood stabilizers may act to restore the balance among aberrant signalling pathways in specific areas of the brain and prevent degeneration.


Subject(s)
Affect/drug effects , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Bipolar Disorder/drug therapy , Brain/drug effects , Lithium Carbonate/pharmacology , Lithium Carbonate/therapeutic use , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Apoptosis/drug effects , Cyclic AMP/metabolism , Humans
10.
Eur Neuropsychopharmacol ; 12(5): 461-75, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12208564

ABSTRACT

Depression is a common disorder that impacts on all aspects of a person's life. For the past 10 years, clinicians have focused on serotonin in their treatment of depression. This is largely due to the growing acceptance of the efficacy and safety of the selective serotonin reuptake inhibitors (SSRIs) in comparison with older tricyclic antidepressants (TCAs). However, evidence for a role of noradrenaline in depression has been accumulating for some time, beginning with the discovery that drugs which either caused or alleviated depression acted to alter noradrenaline metabolism. Until recently, the role of noradrenaline in depression was predicted from clinical experience with noradrenergic TCAs (desipramine, nortriptyline and protriptyline) and selective serotonin and noradrenaline reuptake inhibitors (venlafaxine, milnacipran). The licensing of reboxetine, a selective noradrenaline reuptake inhibitor now allows the role of noradrenaline in depression to be investigated directly. This review presents key data from the literature that support a role for noradrenaline in depression taking into account neurophysiology, psychopharmacology and clinical trial data.


Subject(s)
Depression/drug therapy , Norepinephrine/metabolism , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adrenergic Uptake Inhibitors/therapeutic use , Animals , Antidepressive Agents/therapeutic use , Catechol O-Methyltransferase/genetics , Depression/genetics , Depression/metabolism , Humans , Imipramine/therapeutic use , Monoamine Oxidase/genetics , Morpholines/therapeutic use , Norepinephrine/physiology , Reboxetine , Receptors, Adrenergic/drug effects , Receptors, Adrenergic/metabolism , Treatment Outcome , Tyrosine 3-Monooxygenase/genetics
11.
Biol Psychiatry ; 50(2): 117-22, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11526992

ABSTRACT

BACKGROUND: A disturbance in glutamate neurotransmission has been hypothesized in schizophrenia. Hence, the beneficial effects of pharmacological treatment may be related to adaptive changes taking place in this neurotransmitter system. METHODS: In this study, we investigated the modulation of ionotropic and metabotropic glutamate receptors in the rat brain following acute or chronic exposure to the novel antipsychotic olanzapine. RESULTS: In accordance with the clear distinction between classical and atypical drugs, olanzapine did not alter glutamate receptor expression in striatum. Chronic, not acute, exposure to olanzapine was capable of up-regulating hippocampal mRNA levels for GluR-B and GluR-C, two alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid (AMPA)-forming subunits. This effect could be relevant for the improvement of schizophrenic alterations, which are thought to depend on dysfunction of the glutamatergic transmission within the hippocampal formation. We also found that the expression of group II glutamate metabotropic receptors was up-regulated in the frontal cortex after chronic exposure to clozapine, and to a lesser extent olanzapine, but not with haloperidol. CONCLUSIONS: The adaptive mechanisms taking place in glutamatergic transmission might prove useful in ameliorating some of the dysfunction observed in the brain of schizophrenic patients.


Subject(s)
Antipsychotic Agents/pharmacology , Pirenzepine/analogs & derivatives , Pirenzepine/pharmacology , Receptors, Glutamate/drug effects , Animals , Antipsychotic Agents/administration & dosage , Benzodiazepines , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Olanzapine , Pirenzepine/administration & dosage , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
12.
Pharmacol Ther ; 89(2): 149-70, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11316518

ABSTRACT

It is currently a widely accepted opinion that adaptive, plastic changes in the molecular and cellular components of neuronal signaling systems correlate with the effects on mood and cognition observed after long-term treatment with antidepressant drugs. Protein phosphorylation represents a key step for most signaling systems, and it is involved in the regulation of virtually all cellular functions. Two serine/threonine kinases, Ca2+ /calmodulin-dependent protein kinase II and cyclic AMP-dependent protein kinase, have been shown to be activated in the brain following antidepressant treatment. The changes in kinase activity are mirrored by changes in the phosphorylation of selected protein substrates in subcellular compartments (presynaptic terminals and microtubules), which, in turn, may contribute to the modulation of synaptic transmission observed with antidepressants. The molecular consequences of protein kinase activation may account for some of the alterations in neural function induced by antidepressants, and may suggest novel possible strategies of pharmacological intervention.


Subject(s)
Antidepressive Agents/pharmacology , Calcium-Binding Proteins , Drug Delivery Systems , Mood Disorders/drug therapy , Mood Disorders/physiopathology , Protein Serine-Threonine Kinases/drug effects , Animals , Antidepressive Agents/therapeutic use , Brain Chemistry/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Humans , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity , Phosphorylation , Presynaptic Terminals/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Synaptotagmins
13.
Neuropsychobiology ; 43(3): 150-62, 2001.
Article in English | MEDLINE | ID: mdl-11287794

ABSTRACT

Epidemiological studies clearly indicate that posttraumatic stress disorder (PTSD) is becoming a major health concern worldwide even if still poorly recognized and not well treated. PTSD commonly co-occurs with other psychiatric disorders, and several symptoms overlap with major depressive disorders, anxiety disorders and substance abuse; this may contribute to diagnostic confusion and underdiagnosis. This anxiety disorder provokes significant occupational, psychiatric, medical and psychosocial disability, and its consequences are enormously costly, not only to the survivors and their families, but also to the health care system and society. Work impairment associated with PTSD is very similar to the amount of work impairment associated with major depression. The pathophysiology of PTSD is multifactorial and involves dysregulation of the serotonergic as well as the noradrenergic system. A rational therapeutic approach should normalize the specific psychobiological alterations associated with PTSD. This can be achieved through the use of antidepressant drugs, mainly of those that potentiate serotonergic mechanisms. Recent double-blind placebo-controlled studies report the efficacy of selective serotonin reuptake inhibitors. Several cognitive-behavioral and psychosocial treatments have also been reported to be efficacious and could be considered when treating PTSD patients.


Subject(s)
Antidepressive Agents/therapeutic use , Stress Disorders, Post-Traumatic , Clinical Trials as Topic , Comorbidity , Cost-Benefit Analysis , Diagnosis, Differential , Drug Costs , Humans , Incidence , Psychiatric Status Rating Scales , Psychotherapy , Quality of Life , Receptors, Serotonin/physiology , Risk Factors , Social Behavior , Social Conditions , Stress Disorders, Post-Traumatic/diagnosis , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/pathology , Stress Disorders, Post-Traumatic/psychology
14.
Neuropharmacology ; 40(3): 448-56, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11166338

ABSTRACT

Previous studies showed that the type II Ca(2+)/calmodulin- and cAMP-dependent protein kinases (CaMKII and PKA) are affected by long-term antidepressant treatment in presynaptic and somatodendritic compartments, respectively. This study describes the long-term effects of the selective noradrenaline reuptake inhibitor reboxetine on PKA and CaMKII, in both the microtubule and subsynaptosomal fractions of rat brain. Unlike other antidepressants, chronic reboxetine induced in the cerebrocortical soluble and microtubule fractions a decrease in the [(32)P]cAMP binding to the type II PKA regulatory subunit. No change in the cAMP-dependent endogenous phosphorylation of the protein substrate, microtubule-associated protein 2 was observed. In the hippocampal subsynaptosomal fractions (synaptic vesicles and synaptosomal membranes) reboxetine induced a robust increase in the activity but not in the expression of CaMKII. An increase in the calcium/calmodulin-dependent phosphorylation of presynaptic substrates was also detected. These findings showed that reboxetine modulates post-receptor signal transduction systems in rat brain.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Brain/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Morpholines/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Blotting, Western , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/analysis , Carrier Proteins , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cyclic AMP Receptor Protein/metabolism , Cyclic AMP-Dependent Protein Kinase Type II , Drug Administration Schedule , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/enzymology , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Reboxetine , Subcellular Fractions/metabolism , Synaptic Vesicles/metabolism , Synaptosomes/chemistry , Synaptosomes/enzymology
15.
Neuropsychobiology ; 42(3): 107-19, 2000.
Article in English | MEDLINE | ID: mdl-11015028

ABSTRACT

A wide range of studies have been published over the past two decades that involve the intersection of sleep EEG, insomnia, psychiatric illness (especially depressive disorders) and psychopharmacology. Much of value has been discovered, but there have also been false starts and contradictory results. There is in fact strong evidence that insomnia is associated with medical and psychiatric illness and that the sleepiness associated with insomnia is the cause of many accidents. Thus, the direct (visits to doctors, cost of sleeping medication, complications from use of these medications) and indirect (accidents, quality of life) costs of insomnia are enormous and constitute a major public health problem in the industrialized countries. Believing that it is now timely to assess the state of this important research area, a consensus conference was convened on June 26-28, 1998, in Porto Cervo (Italy) to attempt to clarify the important issues and findings on the clinical effect of the different classes of antidepressant drugs on sleep quality in depression. The participants' consensus on some of the main topics is presented with the hope that this discussion and analysis will contribute to productive research in this important field.


Subject(s)
Antidepressive Agents/therapeutic use , Cost of Illness , Depressive Disorder/drug therapy , Hypnotics and Sedatives/therapeutic use , Sleep Initiation and Maintenance Disorders/drug therapy , Antidepressive Agents/adverse effects , Antidepressive Agents/economics , Cost-Benefit Analysis , Depressive Disorder/economics , Humans , Hypnotics and Sedatives/adverse effects , Hypnotics and Sedatives/economics , Italy , Sleep Initiation and Maintenance Disorders/economics , Sleep Stages/drug effects
16.
J Affect Disord ; 60(1): 61-74, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10940449

ABSTRACT

Social phobia is a common disorder associated with significant psychosocial impairment, representing a substantial public health problem largely determined by the high prevalence, and the lifelong chronicity. Social phobia starts in early childhood or adolescence and is often comorbid with depression, other anxiety disorders, alcohol and substance abuse or eating disorders. This cascade of comorbidity, usually secondary to social phobia, increases the disability associated with the condition. The possibility that social phobia may be a trigger for later developing comorbid disorders directs attention to the need for early effective treatment as a preventive measure. The most recent drug class to be investigated for the psychopharmacological treatment of social phobia is the SSRI group for which there is growing support. The other drug classes that have been evaluated are monoamine oxidase inhibitors (MAOIs), benzodiazepines, and beta-blockers. The SSRIs represent a new and attractive therapeutic choice for patients with generalized social phobia. Recently the first, large scale, placebo-controlled study to assess the efficacy of drug treatment in generalized social phobia has been completed with paroxetine. Paroxetine was more effective in reducing the symptoms than placebo and was well tolerated. Many now regard SSRIs as the drugs of choice in social phobia because of their effectiveness and because they avoid the problems of treatment with benzodiazepines or classical MAOIs.


Subject(s)
Anxiety Disorders/complications , Brain/metabolism , Depressive Disorder, Major/complications , Phobic Disorders , Receptors, Serotonin/metabolism , Adrenergic beta-Antagonists/therapeutic use , Anxiety Disorders/epidemiology , Benzodiazepines/therapeutic use , Cognitive Behavioral Therapy/methods , Comorbidity , Depressive Disorder, Major/epidemiology , Humans , Monoamine Oxidase Inhibitors/therapeutic use , Phobic Disorders/complications , Phobic Disorders/diagnosis , Phobic Disorders/epidemiology , Phobic Disorders/therapy , Prevalence , Psychiatric Status Rating Scales , Selective Serotonin Reuptake Inhibitors/therapeutic use
17.
J Neurochem ; 74(1): 21-33, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617102

ABSTRACT

Depression has been treated pharmacologically for over three decades, but the views regarding the mechanism of action of antidepressant drugs have registered recently a major change. It was increasingly appreciated that adaptive changes in postreceptor signaling pathways, rather than primary action of drugs on monoamine transporters, metabolic enzymes, and receptors, are connected to therapeutic effect. For some of the various signaling pathways affected by antidepressant treatment, it was shown that protein phosphorylation, which represents an obligate step for most pathways, is markedly affected by long-term treatment. Changes were reported to be induced in the function of protein kinase C, cyclic AMP-dependent protein kinase, and calcium/calmodulin-dependent protein kinase. For two of these kinases (cyclic AMP- and calcium/calmodulin-dependent), the changes have been studied in isolated neuronal compartments (microtubules and presynaptic terminals). Antidepressant treatment activates the two kinases and increases the endogenous phosphorylation of selected substrates (microtubule-associated protein 2 and synaptotagmin). These modifications may be partly responsible for the changes induced by antidepressants in neurotransmission. The changes in protein phosphorylation induced by long-term antidepressant treatment may contribute to explain the therapeutic action of antidepressants and suggest new strategies of pharmacological intervention.


Subject(s)
Antidepressive Agents/pharmacology , Brain/enzymology , Protein Kinases/metabolism , Second Messenger Systems/physiology , Animals , Cyclic AMP/physiology , Enzyme Activation , Humans , Neurons/physiology , Phosphoproteins/metabolism , Synapses/metabolism , Synaptic Transmission/drug effects
18.
Int Clin Psychopharmacol ; 14 Suppl 1: S3-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10468322

ABSTRACT

The monoamine hypothesis of depression proposes that the biological basis for depression is a deficiency in the neurotransmitters serotonin or noradrenaline, or both. Over the past 30 years this hypothesis has been refined as more experimental and clinical evidence has emerged. The selective serotonin reuptake inhibitors in particular have made a significant contribution to our understanding of the role of serotonin in depression. Our knowledge of the noradrenergic system is less complete, mainly because, until recently, no equivalent agents were available for this system. With the advent of reboxetine, the first selective noradrenaline reuptake inhibitor, attention is again focusing on the role of noradrenaline in depression. To an extent, the action of the selective inhibitors can be predicted through knowledge of the neuroanatomy of the central and peripheral nervous systems. With regard to depression, the most important pathways are those of the serotonergic and noradrenergic neurones projecting to the prefrontal cortex, from the raphe nucleus and locus coeruleus, respectively. However, increasing the levels of the monoamines in the central nervous system affects many other pathways and a wide range of serotonin and adrenergic receptors, leading to a multiplicity of potential beneficial and adverse events. In addition, the complex intracellular responses are beginning to be examined, again with the aid of the selective antidepressants.


Subject(s)
Brain/physiology , Neurotransmitter Agents/physiology , Norepinephrine/physiology , Serotonin/physiology , Animals , Humans
19.
Neuropsychopharmacology ; 21(2): 211-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10432469

ABSTRACT

The interplay between dopamine and glutamate appears to be relevant in the etiopathology of schizophrenia. Although currently used antipsychotics do not interact with glutamatergic receptors, previous results have demonstrated that the expression profile of ionotropic glutamate receptors can be regulated by drugs such as haloperidol or clozapine. In the present investigation, the mRNA levels for NMDA and AMPA receptor subunits were measured after chronic treatment with the novel antipsychotic agent Seroquel (quetiapine fumarate, quetiapine) as compared to haloperidol and clozapine. Similarly to the prototype atypical clozapine, quetiapine reduced the mRNA expression for NR-1 and NR-2C, two NMDA forming subunits, in the nucleus accumbens. Furthermore, quetiapine, but not haloperidol or clozapine, increased the hippocampal expression for the AMPA subunits GluR-B and GluR-C. The differences between classical and atypical antipsychotics, as well as among the novel agents, might be relevant for specific aspects of their therapeutic activity and could provide valuable information for the role of glutamate in specific symptoms of schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Brain/metabolism , Dibenzothiazepines/pharmacology , Gene Expression Regulation/drug effects , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Transcription, Genetic/drug effects , Animals , Clozapine/pharmacology , Corpus Striatum/metabolism , Haloperidol/pharmacology , Hippocampus/metabolism , Male , Nucleus Accumbens/metabolism , Quetiapine Fumarate , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
20.
J Affect Disord ; 52(1-3): 275-90, 1999.
Article in English | MEDLINE | ID: mdl-10357046

ABSTRACT

Dysthymia, as defined in the American Psychiatric Association and International Classification of Mental Disorders, refers to a prevalent form of subthreshold depressive pathology with gloominess, anhedonia, low drive and energy, low self-esteem and pessimistic outlook. Although comorbidity with panic, social phobic, and alcohol use disorders has been described, the most significant association is with major depressive episodes. Family history is loaded with affective, including bipolar, disorders. The latter finding explains why dysthymia, especially when onset is in childhood, can lead to hypomanic switches, both spontaneously and upon pharmacologic challenge in as many as 30%. Indeed, antidepressants from different classes -tricyclic antidepressants (TCAs), monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase A (RIMAs), selective serotonin-reuptake inhibitors (SSRIs) and, more recently, amisulpride, and spanning noradrenergic, serotonergic as well as dopaminergic mechanisms of action - have been shown to be effective against dysthymia in an average of 65% of cases. This is a promising development because social and characterologic disturbances so pervasive in dysthymia often, though not always, recede with continued pharmacotherapy beyond acute treatment. Despite symptomatic overlap of dysthymia with chronic fatigue syndrome - especially with respect to the cluster of symptoms consisting of low drive, lethargy, lassitude and poor concentration - neither the psychopathologic status, nor the pharmacologic response profile of the latter syndrome is presently understood. Chronic fatigue today is where dysthymia was two decades ago. We submit that the basic science - clinical paradigm that has proven so successful in dysthymia could, before too long, crack down the conundrum of chronic fatigue as well. At a more practical level, we raise the possibility that a subgroup within the chronic fatigue group represents a variant of dysthymia.


Subject(s)
Depressive Disorder, Major/complications , Dysthymic Disorder/complications , Fatigue Syndrome, Chronic/complications , Antidepressive Agents/blood , Antidepressive Agents/therapeutic use , Circadian Rhythm/physiology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/psychology , Dopamine/physiology , Dose-Response Relationship, Drug , Dysthymic Disorder/drug therapy , Dysthymic Disorder/psychology , Family/psychology , Fatigue Syndrome, Chronic/diagnosis , Humans , Hydrocortisone/blood , Self Concept
SELECTION OF CITATIONS
SEARCH DETAIL
...