Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 246
Filter
1.
Cardiovasc Diabetol ; 19(1): 187, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33143700

ABSTRACT

BACKGROUND: Depression and cardiovascular disease (CVD) are among the most common causes of disability in high-income countries, depression being associated with a 30% increased risk of future CV events. Depression is twice as common in people with diabetes and is associated with a 60% rise in the incidence of type 2 diabetes, an independent CVD risk factor. Proprotein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of low-density lipoprotein cholesterol, has been related to a large number of CV risk factors, including insulin resistance. Aim of this study was to investigate whether the presence of depression could affect PCSK9 levels in a population of obese subjects susceptible to depressive symptoms and how these changes may mediate a pre-diabetic risk. RESULTS: In 389 obese individuals, the Beck Depression Inventory (BDI-II) was significantly associated with PCSK9 levels. For every one-unit increment in BDI-II score, PCSK9 rose by 1.85 ng/mL. Depression was associated also with the HOMA-IR (homeostatic model assessment index of insulin resistance), 11% of this effect operating indirectly via PCSK9. CONCLUSIONS: This study indicates a possible mechanism linking depression and insulin resistance, a well-known CV risk factor, providing evidence for a significant role of PCSK9.


Subject(s)
Affect , Cardiovascular Diseases/etiology , Depression/etiology , Insulin Resistance , Obesity/complications , Proprotein Convertase 9/blood , Adult , Biomarkers , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Cross-Sectional Studies , Depression/blood , Depression/diagnosis , Depression/psychology , Female , Heart Disease Risk Factors , Humans , Male , Middle Aged , Obesity/blood , Obesity/diagnosis , Obesity/physiopathology , Retrospective Studies , Risk Assessment
2.
Pharmacol Rev ; 70(3): 475-504, 2018 07.
Article in English | MEDLINE | ID: mdl-29884653

ABSTRACT

Major depressive disorder is one of the most prevalent and life-threatening forms of mental illnesses and a major cause of morbidity worldwide. Currently available antidepressants are effective for most patients, although around 30% are considered treatment resistant (TRD), a condition that is associated with a significant impairment of cognitive function and poor quality of life. In this respect, the identification of the molecular mechanisms contributing to TRD represents an essential step for the design of novel and more efficacious drugs able to modify the clinical course of this disorder and increase remission rates in clinical practice. New insights into the neurobiology of TRD have shed light on the role of a number of different mechanisms, including the glutamatergic system, immune/inflammatory systems, neurotrophin function, and epigenetics. Advances in drug discovery processes in TRD have also influenced the classification of antidepressant drugs and novel classifications are available, such as the neuroscience-based nomenclature that can incorporate such advances in drug development for TRD. This review aims to provide an up-to-date description of key mechanisms in TRD and describe current therapeutic strategies for TRD before examining novel approaches that may ultimately address important neurobiological mechanisms not targeted by currently available antidepressants. All in all, we suggest that drug targeting different neurobiological systems should be able to restore normal function but must also promote resilience to reduce the long-term vulnerability to recurrent depressive episodes.


Subject(s)
Depressive Disorder, Treatment-Resistant , Animals , Antidepressive Agents/classification , Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/etiology , Depressive Disorder, Treatment-Resistant/therapy , Drug Discovery , Humans , Phenotype
3.
Pharmacol Res ; 115: 14-24, 2017 01.
Article in English | MEDLINE | ID: mdl-27838511

ABSTRACT

Nowadays, the goal in the management of type 2 diabetes mellitus (T2DM) remains personalized control of glucose. Since less than 50% of patients with T2DM achieve glycemic treatment goal and most of them take medications for comorbidities associated to T2DM, drug interactions, namely pharmacokinetic and pharmacodynamic interactions, may enhance or reduce the effect of compounds involved in hyperglycemia. Hence, clinicians should be aware of the severe complications in T2DM patients in case of a concomitant use of these medications. It is within this context that this review aims to evaluate the effect of a second drug on the pharmacokinetic of these compounds which may lead, along with several pharmacodynamic interactions, to severe clinical complications, i.e., hypoglycemia. Available drugs already approved in Europe, USA and Japan have been included.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Drug Interactions/physiology , Humans , Hypoglycemic Agents/pharmacokinetics , Insulin/therapeutic use
5.
Mol Psychiatry ; 19(4): 433-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24535456

ABSTRACT

Stress and glucocorticoids alter glutamatergic transmission, and the outcome of stress may range from plasticity enhancing effects to noxious, maladaptive changes. We have previously demonstrated that acute stress rapidly increases glutamate release in prefrontal and frontal cortex via glucocorticoid receptor and accumulation of presynaptic SNARE complex. Here we compared the ex vivo effects of acute stress on glutamate release with those of in vitro application of corticosterone, to analyze whether acute effect of stress on glutamatergic transmission is mediated by local synaptic action of corticosterone. We found that acute stress increases both the readily releasable pool (RRP) of vesicles and depolarization-evoked glutamate release, while application in vitro of corticosterone rapidly increases the RRP, an effect dependent on synaptic receptors for the hormone, but does not induce glutamate release for up to 20 min. These findings indicate that corticosterone mediates the enhancement of glutamate release induced by acute stress, and the rapid non-genomic action of the hormone is necessary but not sufficient for this effect.


Subject(s)
Corticosterone/metabolism , Frontal Lobe/pathology , Neurons/pathology , Presynaptic Terminals/metabolism , Stress, Psychological/pathology , Analysis of Variance , Animals , Aspartic Acid/metabolism , Corticosterone/pharmacology , Dose-Response Relationship, Drug , Electroshock/adverse effects , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Male , Neurons/metabolism , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stress, Psychological/etiology , Synapsins/metabolism , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Synaptosomes/metabolism , Tritium/pharmacokinetics
6.
Int J Neuropsychopharmacol ; 17(5): 779-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24451568

ABSTRACT

Compelling evidence has shown that the effects of antidepressants, increasing extracellular serotonin and noradrenaline as a primary mechanism of action, involve neuroplastic and neurotrophic mechanisms. Brain-derived neurotrophic factor (BDNF) has been shown to play a key role in neuroplasticity and synaptic function, as well as in the pathophysiology of neuropsychiatric disorders and the mechanism of action of antidepressants. The expression of BDNF is mediated by the transcription of different mRNAs derived by the splicing of one of the eight 5' non-coding exons with the 3' coding exon (in rats). The transcription of each non-coding exon is driven by unique and different promoters. We generated a gene reporter system based on hippocampal and cortical neuronal cultures, in which the transcription of luciferase is regulated by BDNF promoters I, II, IV or by cAMP response element (CRE), to investigate the activation of selected promoters induced by monoaminergic antidepressants and by serotonin or noradrenaline agonists. We found that incubation with fluoxetine or reboxetine failed to induce any activation of BDNF promoters or CRE. On the other hand, the incubation of cultures with selective agonists of serotonin or noradrenaline receptors induced a specific and distinct profile of activation of BDNF promoters I, II, IV and CRE, suggesting that the monoaminergic input, absent in dissociated cultures, is essential for the modulation of BDNF expression. In summary, we applied a rapidly detectable and highly sensitive reporter gene assay to characterize the selective activation profile of BDNF and CRE promoters, through specific and different pharmacological stimuli.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Norepinephrine/metabolism , Promoter Regions, Genetic , Serotonin/metabolism , Transcriptional Activation/physiology , Adrenergic Agonists/pharmacology , Animals , Antidepressive Agents/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Fluoxetine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Lithium Carbonate/pharmacology , Morpholines/pharmacology , Neurons/drug effects , Neurons/metabolism , Rats , Reboxetine , Serotonin Receptor Agonists/pharmacology , Transcriptional Activation/drug effects
7.
Neuroscience ; 250: 222-31, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23872394

ABSTRACT

Although extensive evidence demonstrates that repeated administration of amphetamine (AMPH) induces behavioral and neurochemical sensitization, the influence of the developmental timing of AMPH administration is unknown. This is an important issue to address because it could help clarify the influence of early drug exposure on neuronal plasticity and the involvement of dopaminergic sensitization in the etiopathology of neuropsychiatric disorders. Thus, we decided to investigate the molecular alterations induced by the administration of AMPH during adolescence, when repeated exposure to the psychostimulant may interfere with developmental neuroplasticity. We investigated the expression of the neurotrophin brain-derived neurotrophic factor (BDNF) and of two inducible-early genes (arc and cfos) that bridge neuronal activity with long-lasting functional alterations. We found that peri-pubertal treatment with AMPH induces long-lasting changes in the expression of bdnf and of activity-regulated genes in the hippocampus and in the prefrontal/frontal cortex, and leads to alterations of their short-term modulation in response to a subsequent acute AMPH challenge. These data suggest that AMPH exposure in peri-puberty may negatively affect the maturation of brain structures, such as the prefrontal cortex, which facilitate the development of dopamine sensitization and may contribute to dopamine-dependent behavioral dysfunctions and molecular alterations in adulthood.


Subject(s)
Amphetamine , Central Nervous System Stimulants , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Substance Withdrawal Syndrome/genetics , Animals , Blotting, Western , Brain Chemistry/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/physiology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Gene Expression/drug effects , Gene Expression/physiology , Genes, Immediate-Early/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Prefrontal Cortex/metabolism , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Synaptic Transmission/genetics , Synaptic Transmission/physiology
8.
Psychopharmacology (Berl) ; 226(1): 101-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23093383

ABSTRACT

RATIONALE: Combinatory therapy is widely used in psychiatry owing to the possibility that drugs with different mechanisms of action may synergize to improve functions deteriorated in schizophrenia, bipolar disorders, and major depression. While combinatory strategies rely on receptor and synaptic mechanisms, it should also be considered that two drugs may also "interact" on the long-term to determine more robust changes in neuronal plasticity, which represents a downstream target important for functional recovery. OBJECTIVE: The aim of the study is to investigate neuroadaptive changes set in motion by chronic concomitant administration of the novel antipsychotic lurasidone and the mood stabilizer valproate. METHODS: Animals were chronically treated with lurasidone, valproate, or the combination of the two drugs and killed 24 h after the last injection to evaluate alterations of different measures of neuronal plasticity such as the neurotrophin brain-derived neurotrophic factor (BDNF), the immediate early gene Activity-regulated cytoskeletal associated protein, and the epigenetic regulators HDAC 1, 2, and 5 in dorsal and ventral hippocampus. RESULTS: The results suggest that coadministration of lurasidone and valproate produces, when compared to the single drugs, a larger increase in the expression of BDNF in the ventral hippocampus, through the regulation of specific neurotrophin transcripts. We also found that the histone deacetylases were regulated by the drug combination, suggesting that some of the transcriptional changes may be sustained by epigenetic mechanisms. CONCLUSIONS: Our results suggest that the beneficial effects associated with combinatory treatment between a second-generation antipsychotic and a mood stabilizer could result from the ability to modulate neuroplastic molecules, whose expression and function is deteriorated in different psychiatric conditions.


Subject(s)
Affect/drug effects , Antipsychotic Agents/pharmacology , Isoindoles/pharmacology , Neuronal Plasticity/drug effects , Thiazoles/pharmacology , Valproic Acid/pharmacology , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/therapeutic use , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Drug Therapy, Combination , Gene Expression , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Isoindoles/administration & dosage , Isoindoles/therapeutic use , Lurasidone Hydrochloride , Male , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Thiazoles/administration & dosage , Thiazoles/therapeutic use , Valproic Acid/administration & dosage , Valproic Acid/therapeutic use
9.
J Psychopharmacol ; 24(4): 595-603, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19074532

ABSTRACT

Increasing evidence suggests that depression is characterised by impaired brain plasticity that might originate from the interaction between genetic and environmental risk factors. Hence, the aim of this study was to investigate changes in neuroplasticity following exposure to stress, an environmental condition highly relevant to psychiatric disorders, in glucocorticoid receptor-deficient mice (GR(+/-)), a genetic model of predisposition to depression. Specifically, we have analysed the neurotrophin brain-derived neurotrophic factor (BDNF) and the immediate-early gene activity-regulated cytoskeletal-associated protein (Arc), two closely related molecules that can contribute to neuroplastic and morphological changes observed in depression. We found a region-specific influence of the GR-genotype on BDNF levels both under basal and stress conditions. Steady-state levels of BDNF mRNA were unchanged in hippocampus while up-regulated in frontal lobe of GR(+/-) mice. Following exposure to an acute stress, increased processing from pro- to mature BDNF was observed in hippocampal synaptosomes of wild-type mice, but not in GR mutants. Furthermore, the stress-dependent modulation of Arc was impaired in the hippocampus of GR(+/-) mice. These results indicate that GR(+/-) mice show overt differences in the stress-induced modulation of neuroplastic proteins, which may contribute to pathologic conditions that may originate following gene x environment interaction.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cytoskeletal Proteins/metabolism , Depression/metabolism , Frontal Lobe/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Receptors, Glucocorticoid/deficiency , Stress, Psychological/metabolism , Adaptation, Psychological , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Cytoskeletal Proteins/genetics , Depression/genetics , Depression/physiopathology , Depression/psychology , Disease Models, Animal , Frontal Lobe/physiopathology , Genotype , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Phenotype , RNA, Messenger/metabolism , Receptors, Glucocorticoid/genetics , Restraint, Physical , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Synaptosomes/metabolism
10.
Neuroscience ; 159(3): 936-9, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19344636

ABSTRACT

Spinal cord injury (SCI) is a devastating event which causes dramatic changes in the everyday life of the patient. We have found that acute SCI reduced BDNF expression selectively in the hippocampus of lesioned rats, a decrease which persists at least 1 week, thus identifying the modulation of the neurotrophin biosynthesis as an important mechanism underlying brain vulnerability to SCI. These data are the first to show that SCI alters hippocampal BDNF expression and identify the neurotrophin as a potential target through which SCI changes brain functions, a notion that might prove useful in understanding the mechanisms underlying brain vulnerability to SCI.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Spinal Cord Injuries/metabolism , Analysis of Variance , Animals , Autoradiography , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Down-Regulation , Fibroblast Growth Factor 2/metabolism , Frontal Lobe/metabolism , GAP-43 Protein/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Laminectomy , Male , Prefrontal Cortex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
11.
Exp Neurol ; 203(2): 472-80, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17070804

ABSTRACT

Chronic cocaine use leads to pronounced alterations in neuronal functions in brain circuits associated with reward. In the present study, we examined in the rat midbrain the effects of acute, subchronic (5 days) and chronic cocaine treatments (14 days) on the gene expression of transcription factors involved in the development and maintenance of dopaminergic neurons. We show that chronic, but not acute or subchronic, cocaine administration downregulates Nurr1 and Pitx3 transcripts whereas En1 transcripts are upregulated. Conversely, Lmx1b and En2 transcripts are not affected by the drug treatment, indicating that the modulation of the midbrain transcription factors analyzed is highly selective. Interestingly, modification of the gene expression for these transcription factors persists in midbrain as long as two weeks after the last drug administration, suggesting that it may account for some of the enduring alterations in midbrain dopaminergic circuits associated with chronic cocaine use.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/physiology , Mesencephalon/cytology , Mesencephalon/physiology , Neurons/physiology , Transcription Factors/biosynthesis , Animals , Blotting, Western , Cocaine/administration & dosage , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dopamine Uptake Inhibitors/administration & dosage , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Immunohistochemistry , LIM-Homeodomain Proteins , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2 , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/metabolism
12.
Pharmacogenomics J ; 6(2): 95-104, 2006.
Article in English | MEDLINE | ID: mdl-16402079

ABSTRACT

Parkinson's disease (PD) is a chronic, neurodegenerative disease with a 1% incidence in the population over 55 years of age. Movement impairments represent undoubtedly the hallmark of the disorder; however, extensive evidence implicates cognitive deficits as concomitant peculiar features. Brain-derived neurotrophic factor (BDNF) colocalizes with dopamine neurons in the substantia nigra, where dopaminergic cell bodies are located, and it has recently garnered attention as a molecule crucial for cognition, a function that is also compromised in PD patients. Thus, due to its colocalization with dopaminergic neurons and its role in cognition, BDNF might possess a dual role in PD, both as a neuroprotective molecule, since its inhibition leads to loss of nigral dopaminergic neurons, and as a neuromodulator, as its enhanced expression ameliorates cognitive processes. In this review, we discuss the mechanism of action of established as well as novel drugs for PD with a particular emphasis to those interfering with BDNF biosynthesis.


Subject(s)
Antiparkinson Agents , Brain-Derived Neurotrophic Factor/metabolism , Parkinson Disease , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Humans , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/metabolism
13.
Pharmacogenomics J ; 6(1): 8-15, 2006.
Article in English | MEDLINE | ID: mdl-16314887

ABSTRACT

Finding an effective treatment for chronic neurodegenerative disorders still represents an unmet goal. There is considerable evidence that such disorders represent a combination of genetic determinants and failure of neuroprotective mechanisms sparking a wider degree of interest in shedding light on the cellular changes responsible for these devastating disorders. Because of their role in survival or differentiation of developing neurons, as well as the recent discovery of their importance in regulating synaptic plasticity during adulthood, neurotrophic factors have been suggested as essential contributors of the etiology of neurodegenerative disorders. Alzheimer's disease (AD) is a complex, chronic, devastating disease that affects a high percentage of the population over 65 years of age. This review will focus on different pharmacological interventions that are currently in use or drugs under development, narrowing the therapeutic agents to those that interfere with the expression of the trophic factor brain-derived neurotrophic factor (BDNF), a molecule playing a pivotal role in synaptic plasticity and cognition. From these findings, it appears clear that BDNF is implicated in the mechanism of action of drugs that improve cognitive deficits in animal models of AD and in AD patients.


Subject(s)
Alzheimer Disease/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/therapeutic use , Cognition/drug effects , Humans , Life Style , Neuroprotective Agents/therapeutic use , Neurotransmitter Agents/therapeutic use
14.
J Neurochem ; 93(6): 1551-60, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935071

ABSTRACT

Accumulating evidence indicates that antidepressants alter intracellular signalling mechanisms resulting in long-term synaptic alterations which probably account for the delay in clinical action of these drugs. Therefore, we investigated the effects of chronic fluoxetine administration on extracellular signal-regulated kinase (ERK) 1 and 2, a group of MAPKs that mediate signal transduction from the cell surface downstream to the nucleus. Our data demonstrate that 3-week fluoxetine treatment resulted in long-lasting reduction of phospho-ERK 1 and 2. Such an effect depends on the length of the treatment given that no changes were observed after a single drug injection or after 2 weeks of treatment and it is region specific, being observed in hippocampus and frontal cortex but not in striatum. Finally, phospho-ERK 1 and 2 were differently modulated within nucleus and cytosol in hippocampus but similarly reduced in the same compartments of the frontal cortex, highlighting the specific subcellular compartmentalization of fluoxetine. Conversely, imipramine did not reduce the hippocampal phosphorylation of both ERK subtypes whereas it selectively increased ERK 1 phosphorylation in the cytosolic compartment of frontal cortex suggesting a drug-specific effect on this intracellular target. These results point to modulation of phosphorylation, rather than altered expression, as the main target in the action of fluoxetine on this pathway. The reduction of ERK 1/2 function herein reported may be associated with the therapeutic effects of fluoxetine in the treatment of depression.


Subject(s)
Brain/drug effects , Fluoxetine/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Brain/metabolism , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cytoplasm/drug effects , Cytoplasm/enzymology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Imipramine/pharmacology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
16.
Neuropsychobiology ; 46(1): 17-21, 2002.
Article in English | MEDLINE | ID: mdl-12207142

ABSTRACT

Experimental and clinical studies suggest an involvement of the opioid neuropeptide system in schizophrenia. In particular, the prodynorphin (PDYN), the precursor of the dynorphin opioid peptides, has been shown to play an important role in several aspects of human mental diseases. Recently, a functional polymorphism in the promoter of PDYN gene has been described. We studied the possible relationship between this polymorphism and schizophrenia and we found no significant difference in allelic and genotype distributions between schizophrenic patients and control subjects. However, we observed a significant interactive effect with the receptor 3 of dopamine gene (DRD3); in particular, the frequency of subjects carrying PDYN allele 3 being also homozygotes for DRD3 Gly allele (of Ser9Gly polymorphism) was significantly greater in patients than controls. We conclude that PDYN gene polymorphism alone does not alter the risk for schizophrenia but, by an epistatic interaction with the Gly allele of DRD3 gene, may contribute to the susceptibility to this disorder.


Subject(s)
Alleles , Enkephalins/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , Protein Precursors/genetics , Receptors, Dopamine D2/genetics , Schizophrenia/genetics , Case-Control Studies , Genetic Variation , Genotype , Glycine/genetics , Humans , Italy , Receptors, Dopamine D3 , Risk Factors , White People/genetics
17.
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
19.
Mol Psychiatry ; 7(6): 609-16, 2002.
Article in English | MEDLINE | ID: mdl-12140784

ABSTRACT

It is well accepted that events that interfere with the normal program of neuronal differentiation and brain maturation may be relevant for the etiology of psychiatric disorders, setting the stage for synaptic disorganization that becomes functional later in life. In order to investigate molecular determinants for these events, we examined the modulation of the neurotrophin brain-derived neurotrophic factor (BDNF) and the glutamate NMDA receptor following 24 h maternal separation (MD) on postnatal day 9. We found that in adulthood the expression of BDNF as well as of NR-2A and NR-2B, two NMDA receptor forming subunits, were significantly reduced in the hippocampus of MD rats whereas, among other structures, a slight reduction of NR-2A and 2B was detected only in prefrontal cortex. These changes were not observed acutely, nor in pre-weaning animals. Furthermore we found that in MD rats the modulation of hippocampal BDNF in response to an acute stress was altered, indicating a persistent functional impairment in its regulation, which may subserve a specific role for coping with challenging situations. We propose that adverse events taking place during brain maturation can modulate the expression of molecular players of cellular plasticity within selected brain regions, thus contributing to permanent alterations in brain function, which might ultimately lead to an increased vulnerability for psychiatric diseases.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Gene Expression Regulation/physiology , Hippocampus/physiology , Maternal Deprivation , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Female , Male , Protein Subunits/genetics , RNA Probes , Rats , Rats, Wistar , Reference Values
20.
Mol Psychiatry ; 7(2): 208-12, 2002.
Article in English | MEDLINE | ID: mdl-11840314

ABSTRACT

Abnormalities in protein kinase A (PKA) and Rap1 have recently been reported in depressed patients. The aim of the present study was to investigate the levels of these proteins in platelets from untreated unipolar and bipolar depressed patients with psychotic features. The levels PKA and Rap1 were assessed by Western blot analysis and immunostaining in 37 drug-free patients and 29 healthy subjects. Both unipolar and bipolar patients with psychotic depression have significantly lower levels of platelet regulatory type I and higher levels of catalytic subunits of PKA than controls, whereas the levels of regulatory type II were higher only in psychotic unipolar patients. No significant differences were found in the immunolabeling of both Rap1 and actin among groups. These findings support the idea that besides nonpsychotic depression, abnormalities of PKA could be linked, albeit in a somewhat different way, with psychotic depression.


Subject(s)
Bipolar Disorder/metabolism , Cyclic AMP/metabolism , Depressive Disorder/metabolism , Signal Transduction/physiology , Adult , Blood Platelets/chemistry , Blood Platelets/enzymology , Blotting, Western , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Male , Middle Aged , rap1 GTP-Binding Proteins/analysis , rap1 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...