Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Biomed Pharmacother ; 154: 113612, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36049313

ABSTRACT

The psychedelic 5-HT2A receptor (5HT2AR) agonist psilocybin (or the active metabolite psilocin) has emerged as potential useful drug for various neuropsychiatric diseases, with a rapid onset of therapeutic activity. However, the mechanisms responsible for such effects remain incompletely characterized. We aimed to study in vitro pharmacological profile and in vivo acute mechanism of psilocin/psilocybin. Competition binding studies with psilocin were performed in brain and cell cultures. The role of 5HT2AR, 5-HT2C receptors (5HT2CR) and 5-HT1A receptors (5HT1AR) on the psychosis-like head-twitch response (HTR) and on body temperature in mice after psilocybin administration were evaluated. Psilocin showed similar affinities for 5HT2AR (Ki: 120-173 nM), 5HT2CR (Ki: 79-311 nM) and 5-HT1AR (Ki: 152-146 nM) in human and mice brain. Psilocybin induced a dose-dependent HTR (maximal effect 17.07 ± 1.31 at 1 mg/kg i.p.) that was completely suppressed by the 5HT2AR antagonist MDL11939 (1 mg/kg). Higher doses of psilocybin (3 mg/kg) induced lower HTR (9.00 ± 0.53). The 5HT2CR antagonist SB242084 (0.1 mg/kg) increased HTR exerted by psilocybin (3 mg/kg). Psilocybin significantly raised core body temperature at low dose (0.125 mg/kg) (Emax=0.67 ± 0.15 °C), whereas a significant decrease was induced by doses over 1 mg/kg (Emax = -1.31 ± 0.16 °C). Pre-treatment with the 5HT1AR antagonist WAY100635 reversed the decrease of body temperature after psilocybin (1 mg/kg), causing hyperthermia (Emax = 0.94 ± 0.26 °C). The present work provides key findings on the 5HT2AR, 5-HT2CR and 5HT1AR involvement in the acute central effects of psilocybin. The results may be relevant for understanding the mechanism of action underlying the therapeutic effects and side effects of this psychedelic drug.


Subject(s)
Hallucinogens , Psilocybin , Animals , Body Temperature Regulation , Hallucinogens/pharmacology , Humans , Mice , Psilocybin/pharmacology , Receptor, Serotonin, 5-HT1A , Serotonin
2.
CNS Drugs ; 32(5): 411-420, 2018 05.
Article in English | MEDLINE | ID: mdl-29736744

ABSTRACT

Ketamine and its enantiomer S-ketamine (esketamine) are promising candidates to produce a rapid-onset antidepressant effect in treatment-resistant depression. Ketamine causes continued blockade of the glutamate N-methyl-D-aspartate (NMDA) receptor, though this might not primarily mediate the antidepressant effect. Alternative hypotheses include selectivity for the NMDA receptor subtype containing the NMDA receptor subunit 2B (NR2B), inhibition of the phosphorylation of the eukaryotic elongation factor 2 (eEF2) kinase, increased expression of brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrKB), and activation of the mammalian target of rapamycin (mTOR) signaling pathway, alongside other independent actions attributed to the ketamine metabolism to R-hydroxynorketamine (R-HNK). The enantiomer S-ketamine (esketamine) displays approximately fourfold greater affinity for the glutamate NMDA receptor in vitro than R-ketamine. Proof-of-concept single-dose and repeat-dose studies with intravenous ketamine show a significant antidepressant and probably antisuicidal effect in the short term, with response rates over 60% as early as 4.5 h after a single dose, with a sustained effect after 24 h, and over 40% after 7 days. This response can be further sustained over several weeks with repeated doses (two to three doses per week). Tolerability seems acceptable in the short term, with transient elevation of blood pressure and mild and transient dissociative and psychotomimetic effects. Intranasal esketamine has shown a comparable antidepressant effect, which has resulted in the US FDA granting the drug a "breakthrough therapy" designation, and theoretically it may offer an improved tolerability profile. However, major concerns remain regarding an effective protocol to maintain the clinical antidepressant effect of ketamine seen with acute administration and the safety of ketamine and esketamine in the long term, specifically related to potential neurocognitive and urologic toxicity, together with the potential induction of substance use disorders. Ketamine and esketamine are not currently approved treatments for depression, but the clinical use of ketamine is increasing in a variety of practice settings internationally.


Subject(s)
Antidepressive Agents/adverse effects , Antidepressive Agents/therapeutic use , Ketamine/adverse effects , Ketamine/therapeutic use , Animals , Antidepressive Agents/pharmacology , Humans , Ketamine/pharmacology
3.
Neuroscience ; 277: 541-51, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25075716

ABSTRACT

Enhanced brain apoptosis (neurons and glia) may be involved in major depression (MD) and schizophrenia (SZ), mainly through the activation of the intrinsic (mitochondrial) apoptotic pathway. In the extrinsic death pathway, pro-apoptotic Fas-associated death domain (FADD) adaptor and its non-apoptotic p-Ser194 FADD form have critical roles interacting with other death regulators such as phosphoprotein enriched in astrocytes of 15 kDa (PEA-15) and extracellular signal-regulated kinase (ERK). The basal status of FADD (protein and messenger RNA (mRNA)) and the effects of psychotropic drugs (detected in blood/urine samples) were first assessed in postmortem prefrontal cortex of MD and SZ subjects (including a non-MD/SZ suicide group). In MD, p-FADD, but not total FADD (and mRNA), was increased (26%, n=24; all MD subjects) as well as p-FADD/FADD ratio (a pro-survival marker) in antidepressant-free MD subjects (50%, n=10). In contrast, cortical FADD (and mRNA), p-FADD, and p-FADD/FADD were not altered in SZ brains (n=21) regardless of antipsychotic medications (except enhanced mRNA in treated subjects). Similar negative results were quantified in the non-MD/SZ suicide group. In MD, the regulation of multifunctional PEA-15 (i.e., p-Ser116 PEA-15 blocks pro-apoptotic FADD and PEA-15 prevents pro-survival ERK action) and the modulation of p-ERK1/2 were also investigated. Cortical p-PEA-15 was not changed whereas PEA-15 was increased mainly in antidepressant-treated subjects (16-20%). Interestingly, cortical p-ERK1/2/ERK1/2 ratio was reduced (33%) in antidepressant-free when compared to antidepressant-treated MD subjects. The neurochemical adaptations of brain FADD (increased p-FADD and pro-survival p-FADD/FADD ratio), as well as its interaction with PEA-15, could play a major role to counteract the known activation of the mitochondrial apoptotic pathway in MD.


Subject(s)
Brain/metabolism , Depressive Disorder, Major/metabolism , Fas-Associated Death Domain Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Phosphoproteins/metabolism , Schizophrenia/metabolism , Adult , Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Apoptosis Regulatory Proteins , Depressive Disorder, Major/drug therapy , Fas-Associated Death Domain Protein/genetics , Female , Humans , Immunoblotting , Male , Middle Aged , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Messenger/metabolism , Schizophrenia/drug therapy , Suicide
4.
Transl Psychiatry ; 3: e221, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23340504

ABSTRACT

Overexpression of the mammalian homolog of the unc-18 gene (munc18-1) has been described in the brain of subjects with schizophrenia. Munc18-1 protein is involved in membrane fusion processes, exocytosis and neurotransmitter release. A transgenic mouse strain that overexpresses the protein isoform munc18-1a in the brain was characterized. This animal displays several schizophrenia-related behaviors, supersensitivity to hallucinogenic drugs and deficits in prepulse inhibition that reverse after antipsychotic treatment. Relevant brain areas (that is, cortex and striatum) exhibit reduced expression of dopamine D(1) receptors and dopamine transporters together with enhanced amphetamine-induced in vivo dopamine release. Magnetic resonance imaging demonstrates decreased gray matter volume in the transgenic animal. In conclusion, the mouse overexpressing brain munc18-1a represents a new valid animal model that resembles functional and structural abnormalities in patients with schizophrenia. The animal could provide valuable insights into phenotypic aspects of this psychiatric disorder.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Munc18 Proteins/metabolism , Schizophrenia/metabolism , Animals , Caspase 3/metabolism , Disease Models, Animal , Dopamine/metabolism , Humans , Lipid Peroxidation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis , Motor Activity/genetics , Munc18 Proteins/genetics , Nerve Fibers, Unmyelinated/pathology , Organ Size/genetics , Phenotype , Recognition, Psychology/physiology , Reflex, Startle/genetics , SNARE Proteins/metabolism , Social Behavior
5.
Psychopharmacology (Berl) ; 226(1): 177-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23093381

ABSTRACT

RATIONALE: Regulator of G-protein signaling (RGS) proteins, RGS4 and RGS10, may be involved in the pathophysiology of schizophrenia. RGS4 has attracted special interest since the reports of genetic association between SNPs in RGS4 and schizophrenia. However, there is no information about the subcellular distribution of RGS4 and RGS10 proteins in psychiatric disorders. OBJECTIVES: Plasma membrane RGS4 and cytosolic RGS10 protein immunoreactivity in prefrontal cortex from schizophrenic subjects (n = 25), non-diagnosed suicides (n = 13), and control subjects (n = 35), matched by age, gender, and postmortem delay, was analyzed by western blot. A second group of depressed subjects (n = 25) and control subjects (n = 25) was evaluated. The effect of the antipsychotic or antidepressant treatments was also assessed. RESULTS: No significant differences in plasma membrane RGS4 and cytosolic RGS10 protein expression were observed between schizophrenic subjects, non-diagnosed suicides, and control subjects. However, RGS4 immunoreactivity was significantly higher (Δ = 33 ± 10 %, p < 0.05) in the antipsychotic-treated subgroup (n = 12) than in the antipsychotic-free subgroup (n = 13). Immunodensities of plasma membrane RGS4 and cytosolic RGS10 proteins did not differ between depressed and matched control subjects. CONCLUSIONS: Expression of RGS4 and RGS10 proteins at their predominant subcellular location was studied in the postmortem brain of subjects with psychiatric disorders. The results suggest unaltered membrane RGS4 and cytosolic RGS10 proteins levels in schizophrenia and major depression. Antipsychotic treatment seems to increase membrane RGS4 immunoreactivity. Further studies are needed to elucidate RGS4 and RGS10 functional status.


Subject(s)
Antipsychotic Agents/therapeutic use , Depressive Disorder, Major/metabolism , Prefrontal Cortex/metabolism , RGS Proteins/biosynthesis , Schizophrenia/metabolism , Adult , Antipsychotic Agents/blood , Biopsy , Blotting, Western , Cell Membrane/metabolism , Cytosol/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/pathology , Female , Humans , Male , Middle Aged , Postmortem Changes , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Schizophrenia/drug therapy , Schizophrenia/pathology , Suicide
6.
Biochem Pharmacol ; 83(2): 260-8, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22093909

ABSTRACT

Rimonabant (SR141716) was the first potent and selective cannabinoid CB1 receptor antagonist synthesized. Several data support that rimonabant behaves as an inverse agonist. Moreover, there is evidence suggesting that this inverse agonism may be CB1 receptor-independent. The aim of the present study was to elucidate whether the effect of rimonabant over G protein activation in postmortem human brain is CB1 dependent or independent. [(35)S]GTPγS binding assays and antibody-capture [(35)S]GTPγS scintillation proximity assays (SPA) were performed in human and mice brain. [(3)H]SR141716 binding characteristics were also studied. Rimonabant concentration-dependently decreased basal [(35)S]GTPγS binding to human cortical membranes. This effect did not change in the presence of either the CB1 receptor agonist WIN 55,212-2, the CB1 receptor neutral antagonist O-2050, or the CB1 allosteric modulator Org 27569. [(35)S]GTPγS binding assays performed in CB1 knockout mice brains revealed that rimonabant inhibited the [(35)S]GTPγS binding in the same manner as it did in wild-type mice. The SPA combined with the use of specific antibody-capture of G(α) specific subunits showed that rimonabant produces its inverse agonist effect through G(i3), G(o) and G(z) subtypes. This effect was not inhibited by the CB1 receptor antagonist O-2050. Finally, [(3)H]SR141716 binding assays in human cortical membranes demonstrated that rimonabant recognizes an additional binding site other than the CB1 receptor orthosteric binding site recognized by O-2050. This study provides new data demonstrating that at least the inverse agonist effect observed with >1µM concentrations of rimonabant in [(35)S]GTPγS binding assays is not mediated by the CB1 receptor in human brain.


Subject(s)
Brain/metabolism , Drug Inverse Agonism , GTP-Binding Proteins/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/agonists , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/physiology , Animals , Brain/drug effects , Dose-Response Relationship, Drug , GTP-Binding Proteins/agonists , Humans , Mice , Mice, Knockout , Piperidines/metabolism , Pyrazoles/metabolism , Rimonabant
7.
Neuroscience ; 196: 1-15, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21925237

ABSTRACT

Cocaine induces apoptotic effects in cultured cells and in the developing brain, but the aberrant activation of cell death in the adult brain remains inconclusive, especially in humans. This postmortem human brain study examined the status of canonical apoptotic pathways, signaling partners, and the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1), a sensor of DNA damage, in prefrontal cortex (PFC) of a small but well-characterized cohort of cocaine abusers (n=10). For comparison, the chosen targets were also quantified in the cerebral cortex of cocaine-treated rats. In the PFC of cocaine abusers, FS7-associated cell surface antigen (Fas) receptor aggregates and Fas-associated death domain (FADD) adaptor were reduced (-26% and -66%, respectively) as well as the content of mitochondrial cytochrome c (-61%). In the same brain samples of cocaine abusers, the proteolytic cleavage of PARP-1 was increased (+39%). Nuclear PARP-1 degradation, possibly a consequence of increased mitochondrial oxidative stress, involved the activation of apoptosis-inducing factor (AIF) and not that of caspase-3. In the PFC of cocaine abusers, several signaling molecules associated with cocaine/dopamine and/or apoptotic pathways were not significantly altered, with the exception of anti-apoptotic truncated DARPP-32 (t-DARPP), a truncated isoform of dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32), whose content was decreased (-28%). Chronic exposure to cocaine in rats, including withdrawal for 3 days, did not alter Fas-FADD receptor complex, cytochrome c, caspase-3/fragments, AIF, PARP-1 cleavage, and associated signaling in the cerebral cortex. Chronic cocaine and abstinence, however, increased the content of t-DARPP (+39% and +47%) in rat brain cortex. The major findings indicate that cocaine addiction in humans is not associated with abnormal activation of extrinsic and intrinsic apoptotic pathways in PFC. The downregulation of Fas-FADD receptor complex and cytochrome c could reflect the induction of contraregulatory adaptations or non-apoptotic (neuroplastic) actions induced by the psychostimulant. The enhanced degradation of nuclear PARP-1, a hallmark of apoptosis, indicates the possibility of aberrant cell death.


Subject(s)
Apoptosis/physiology , Cerebral Cortex/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Prefrontal Cortex/metabolism , Signal Transduction/physiology , Adult , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor/metabolism , Caspase 3/metabolism , Cerebral Cortex/drug effects , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Fas-Associated Death Domain Protein/metabolism , Female , Humans , Male , Middle Aged , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , fas Receptor/metabolism
8.
J Oral Rehabil ; 37(11): 827-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21039747

ABSTRACT

In humans, diurnal tooth-clenching and other oral stereotyped behaviour are associated with stress/anxiety. In rodents, gnawing/biting of objects is observed during exposure to stress. Both nigrostriatal and mesocortical dopaminergic systems are involved in the development of this coping behaviour. To clarify the relationship between central dopaminergic activity and stress-induced parafunctional masticatory behaviour, using microdialysis in vivo, we assessed the changes in extracellular dopamine concentrations in both prefrontal cortex and striatum of rats subjected to a mild tail pinch. The animals were divided into two groups according to the degree of non-functional masticatory activity (NFMA) displayed during exposure to tail pinch. In prefrontal cortex, rats which displayed severe NFMA showed a greater increase in extracellular dopamine concentration in relation to basal values (Emax=184±26%) than those which did not display this coping behaviour (Emax=139± 23%) (F(NFMA) [1,86]=3·97; P<0·05) (n=17). A positive association was also found between cortical dopamine maximal value from baseline and the degree of NFMA displayed (r=0·36; P<0·05) (n=17). There were no significant differences in the tail-pinch-induced striatal dopamine increase between both groups of rats (Emax=130±10%) (n=17). These results provide further evidence in support of prefrontal dopamine playing a relevant role in the expression of stress-induced masticatory coping behaviour.


Subject(s)
Bruxism/physiopathology , Corpus Striatum/metabolism , Dopamine/metabolism , Mastication/physiology , Prefrontal Cortex/metabolism , Stress, Physiological , Analysis of Variance , Animals , Male , Microdialysis , Rats , Rats, Sprague-Dawley
9.
Actas Esp Psiquiatr ; 38(1): 22-32, 2010.
Article in English, Spanish | MEDLINE | ID: mdl-20931407

ABSTRACT

Bipolar disorder is considered an important public health problem in the world. The depressive phase is the most important in terms of frequency, duration, and impairment of the quality of life. Common treatment of bipolar depression usually includes antidepressants, mood stabilizers and antipsychotics in different combinations, despite not having a specific indication for that. Quetiapine is the first drug in Europe that has obtained a specific indication for the treatment of bipolar depression, due to a pharmacologic profile that makes it to act on the three neurotransmitter systems involved in bipolar depression neurobiology. Regarding the dopaminergic pathway, quetiapine leads to an increasing of prefrontal dopamine release by antagonism of5-HT2A receptors, partial agonist of 5-HT1A and antagonism of a2 adrenoceptors. Quetiapine also enhances the serotoninergic transmission by increasing the density of receptors5-HT1A in the prefrontal cortex and by antagonism of 5-HT2A receptors and a2 adrenoceptors. On the other hand, norquetiapine, the main active metabolite of quetiapine, actsas a 5-HT2C antagonist and is a potent inhibitor of norepinephrine transporter (NET). NET inhibition leads to an increase of noerpinephrine in the synapse, and together with the increase of prefrontal dopamine and serotonin, could explain the antidepressive effect demonstrated by quetiapine in several clinical trials. Quetiapine's action on glutamatergicand GABAergic receptors represents an interesting object of research, together with a potential neuroprotective effect that have already been observed in animal models.


Subject(s)
Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Dibenzothiazepines/therapeutic use , Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Dibenzothiazepines/pharmacology , Humans , Neurobiology , Neuroprotective Agents/pharmacology , Quetiapine Fumarate , Receptors, Dopamine/drug effects , Receptors, Serotonin/drug effects
10.
Neuroscience ; 161(1): 23-38, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19303913

ABSTRACT

Fas-associated protein with death domain (FADD) is a multifunctional protein that can induce both apoptotic and non-apoptotic actions. Recently, FADD was found downregulated in the prefrontal cortex of opiate abusers, which suggested an attenuation of Fas death signals in human addicts. Phosphorylation of FADD (Ser194) has been reported to regulate its non-apoptotic activity, which might include the induction of neuroplastic effects in the brain. This postmortem brain study examined the status of phosphorylated (p)-Ser194 FADD and signaling pathways involved in neuroplasticity in the prefrontal cortex (BA 9) of short-term (ST) and long-term (LT) heroin or methadone abusers. In these subjects, the content of monomeric p-FADD was significantly increased when compared with that in age-, gender-, and postmortem delay-matched controls (all addicts: 65%, n=26; ST abuse: 51%; n=11; LT abuse: 75%, n=15). Oligomeric p-FADD forms were modestly increased (11%-23%). At the subcellular level, opiate addiction upregulated the expression of monomeric p-FADD in the nucleus (110%) and that of p-oligomers in the cytosol (66%). In LT opiate addicts (but not ST abusers), a pronounced downregulation of p-extracellular signal-regulated kinase (ERK)1/2 (52%) and p-c-Jun NH(2)-terminal protein kinase (JNK)1/2 (51%), but not p-p38 mitogen-activated protein kinase (MAPK), was quantified in the prefrontal cortex (total homogenate and subcellular compartments). Similarly, the signaling pathway mediated by p-phosphoprotein enriched in astrocytes of 15 kDa (PEA-15) protein and its phosphorylating kinase p-Akt1 was also downregulated in cortical homogenate (43% and 41%, respectively) and cytosolic preparations of chronic opiate addicts. The results indicate that opiate addiction in humans is associated with an altered balance between p-Ser194 FADD (increased) and total FADD (decreased) in brain, which may favor its neuroplastic actions. The interaction between p-FADD (upregulated) and neuronal pathways (downregulated) could play a relevant role in mediating specific forms of structural and behavioral neuroplasticity.


Subject(s)
Fas-Associated Death Domain Protein/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Mitogen-Activated Protein Kinases/physiology , Neuronal Plasticity , Opioid-Related Disorders/metabolism , Phosphoproteins/physiology , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-akt/physiology , Adult , Apoptosis Regulatory Proteins , Female , Humans , Male , Phosphorylation , Serine/metabolism , Signal Transduction , Time Factors
11.
Neuroscience ; 157(1): 105-19, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18834930

ABSTRACT

Opiate addiction is a chronic medical disorder characterized by drug tolerance and dependence, behavioral sensitization, vulnerability to compulsive relapse, and high mortality. In laboratory animals, the potential effect of opiate drugs to induce cell death by apoptosis is a controversial topic. This postmortem human brain study examined the status of the extrinsic and intrinsic apoptotic pathways in the prefrontal cortex of a large group of well-characterized heroin or methadone abusers. In these subjects (n=36), the immunocontent of apoptosis-1 protein (Fas) death receptor did not differ from that in age-, gender-, and postmortem delay-matched controls. In contrast, Fas-associated protein with death domain (FADD), the mediator of the death signal, was significantly decreased in the same brain samples (all addicts: 30%, n=36; short-term abuse (ST): 31%, n=15; long-term abuse (LT): 29%, n=21). The initiator caspase-8 was not altered, but FLIP(L) (Fas-associated protein with death domain-like interleukin-1beta-converting enzyme-inhibitory protein), a dominant inhibitor of caspase-8, was increased in LT addicts (19%). In the intrinsic pathway, the pro-apoptotic mitochondrial proteins Bax (Bcl-2-associated X protein) and AIF (apoptosis-inducing factor) remained unchanged, but cytochrome c was decreased (all addicts: 25%; ST: 31%; LT: 20%) and anti-apoptotic B-cell leukemia 2 (Bcl-2) increased in LT addicts (24%). The content of executioner caspase-3 and the pattern of cleavage of the nuclear enzyme poly-(ADP-ribose)-polymerase-1 (PARP-1) were similar in opiate addicts and control subjects. Taken together, the data revealed that the extrinsic and intrinsic canonical apoptotic pathways are not abnormally activated in the prefrontal cortex of opiate abusers. Instead, the chronic modulation of some of their components (downregulation of FADD and cytochrome c; upregulation of FLIP(L) and Bcl-2) suggests the induction of non-apoptotic actions by opiate drugs related to phenomena of synaptic plasticity in the brain. These neurochemical adaptations could play a major role in the development of opiate tolerance, sensitization and relapse in human addicts.


Subject(s)
Apoptosis/physiology , Opioid-Related Disorders/pathology , Prefrontal Cortex/pathology , Signal Transduction/physiology , Acute Disease , Adult , Aging/metabolism , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Brain Chemistry , Chronic Disease , Female , Hair/chemistry , Humans , Male , Middle Aged , Narcotics/analysis , Narcotics/blood , Neuronal Plasticity/physiology , Opioid-Related Disorders/metabolism , Prefrontal Cortex/metabolism , Sex Characteristics , Synapses/physiology , Young Adult
12.
Genes Brain Behav ; 7(6): 649-58, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18363858

ABSTRACT

Animal models represent a very useful tool for the study of depressive-like behavior and for the evaluation of the therapeutic efficacy of antidepressants. Nevertheless, gene expression patterns of these different animal models and whether genes classically associated with human major depression are present in these genetic profiles remain unknown. Gene expression was evaluated in three animal models of depression: acute treatment with reserpine, olfactory bulbectomy and chronic treatment with corticosterone. Gene expression analysis was carried out using the Affymetrix GeneChip technology. The results were evaluated using the GeneChip Operating software (Gcos 1.3) and analyzed with the GeneSpring GX v7.3 bioinformatics software (Agilent) and dChip 2005 software. Expression changes were validated with quantitative real-time polymerase chain reaction (RT-PCR) assays. Many transcripts were differentially expressed in the cortex of depressed-like animals in each model. Gene ontology analysis showed that significant gene changes were clustered primarily into functional neurochemical pathways associated with apoptosis and neuronal differentiation. When expression profiles were compared among the three models, the number of transcripts differentially expressed decreased and only two transcripts (complement component 3 and fatty acid-binding protein 7) were differentially expressed in common. Both genes were validated with RT-PCR. Moreover, five (Htr2a, Ntrk3, Crhr1, Ntrk2 and Crh) of the genes classically related to human major depression were differentially expressed in at least one of these models. The different animal models of depression share relevant characteristics although gene expression patterns are different among them. Moreover, some of the classical genes related to human major depression are differentially expressed in these models.


Subject(s)
Brain Chemistry/genetics , Cerebral Cortex/metabolism , Depressive Disorder/genetics , Gene Expression Regulation/genetics , Adrenergic Uptake Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cerebral Cortex/physiopathology , Complement C3/genetics , Corticosterone/pharmacology , Denervation , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Fatty Acid-Binding Proteins/genetics , Gene Expression Profiling , Humans , Male , Nerve Tissue Proteins/genetics , Olfactory Bulb/surgery , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology , Species Specificity
13.
Rev Neurol ; 43(8): 476-80, 2006.
Article in Spanish | MEDLINE | ID: mdl-17033981

ABSTRACT

AIM: To present the experimental data that support the hypothesis that the imidazoline I(2) receptors may be assessed as a biological marker to establish diagnosis and grade of human gliomas. DEVELOPMENT: Gliomas constitute the most important group of brain neoplasm in humans. In these tumours accurate histopathologic diagnosis is a first crucial prerequisite for patient treatment. However, current grading schemes are still limited by subjective histologic criteria. Therefore, the search for new molecular and biological markers of gliomas represents a crucial step. In this context, it has been reported a significant increase in I(2) density in human gliomas when compared with normal brain tissue and other intracranial non-glial tumours. Moreover, this increase seems to fit well with the degree of malignancy in human gliomas. Thus, in glioblastomas multiformes the I(2) density is 1.4 times higher than in anaplastic astrocytomas and 2.2 higher than in low-grade astrocytomas. CONCLUSIONS: The present results demonstrate that the measurement of the I(2) density by positron emission tomography techniques could be used in the future for grading and prognosis of human gliomas. This could avoid the current need for tumour biopsies in order to obtain a histopathologic diagnosis.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/chemistry , Brain Neoplasms/diagnosis , Glioma/chemistry , Glioma/diagnosis , Receptors, Drug/analysis , Humans , Imidazoline Receptors
14.
Neuroscience ; 141(1): 357-69, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16697114

ABSTRACT

Knowledge about the noradrenergic system in birds is very scarce even though their biological diversity and complex social behavior make them an excellent model for studying neuronal functions and developmental biology. While the role of norepinephrine has been described in depth in a large number of central and peripheral functions in mammals, reports for avian species are limited. The radioligand [(3)H]RX 821002 ([(3)H]1,4-[6,7(n)3H]-benzodioxan-2-methoxy-2-yl)-2-imidazol) has been used to map and characterize alpha(2)-adrenoceptors through the chicken brain using in vitro autoradiography and membrane homogenates binding assays. [(3)H]RX 821002 showed a saturable and high affinity binding to a site compatible with alpha(2)-adrenoceptor, and to a serotonergic component. The autoradiographic assays displayed a similar alpha(2)-adrenoceptor distribution than those previously reported in birds using other radioligands such as [(3)H]UK 14304 ([(3)H]5-bromo-N-(4,5-dihydro-1H-imidazol-2-yl)-6-quinoxalinamine) or [(3)H]clonidine. [(3)H]RX 821002 binding pharmacological characterization was carried out in different chicken brain regions using membrane homogenates for competition assays with different alpha(2)-adrenoceptor agonists and antagonists drugs (oxymetazoline, BRL 44408 [2-(2H-(1-methyl-1,3-dihydroisoindole)methyl)-4,5-dihydroimidazole] ARC 239 [2-(2-4-(O-methoxyphenyl)-piperazin-1-yl)-ethyl-4,4-dimethyl-1,3-(2H,4H)-isoquinolindione], prazosin, UK 14304 and RX 821002). The results showed alpha(2A) as the predominant alpha(2)-adrenoceptor subtype in the chicken brain while alpha(2B)- and/or alpha(2C)-adrenoceptor subtypes were detected only in the telencephalon. RX 821002, serotonin (5-HT) and 8-OH-DPAT [8-hydroxy-2-(di-n-propylamino)tetralin] competition assays, and competition binding assays performed in the presence of serotonin demonstrated that [(3)H]RX 821002 binds with higher affinity to a serotonergic component, probably 5-HT(1A) receptors, than to the alpha(2)-adrenoceptors. Similar pharmacological properties for the alpha(2)-adrenoceptor component were observed both in rat and chicken brain. The results demonstrate that the different alpha(2)-adrenoceptor subtypes are present in chicken brain and suggest that these receptors are highly conserved through evolution.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Adrenergic alpha-Antagonists/pharmacokinetics , Brain/drug effects , Idazoxan/analogs & derivatives , Adrenergic alpha-Agonists/pharmacology , Animals , Binding, Competitive/drug effects , Brain/anatomy & histology , Chickens , Dose-Response Relationship, Drug , Drug Interactions , Idazoxan/pharmacokinetics , Idazoxan/pharmacology , In Vitro Techniques , Male , Oxymetazoline/pharmacology , Radioligand Assay/methods , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Tritium/pharmacokinetics
15.
Neuroscience ; 140(2): 635-43, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16563642

ABSTRACT

The CB1 cannabinoid receptor (CB1) is the predominant type of cannabinoid receptor in the CNS, in which it displays a unique anatomical distribution and is present at higher densities than most other known seven transmembrane domain receptors. Nevertheless, as with almost all seven transmembrane domain receptors, the tertiary and quaternary structure of this receptor is still unknown. Studies of CB1 in rat cerebral tissue are scarce, and even less is known regarding the expression of CB1 in the human brain. Thus, the aim of the present work was to characterize CB1 expression in membranes from postmortem human brain using specific antisera raised against this protein. Western blot analysis of P1 and P2 fractions, and crude plasma membrane preparations from the prefrontal cortex showed that CB1 migrated as a 60 kDa monomer under reducing conditions. These data were confirmed by blotting experiments carried out with human U373MG astrocytoma cells as a positive control for CB1 expression and wild-type CHO cells as negative control. In addition, when proteins were solubilized in the absence of dithiothreitol, the anti-human CB1 antiserum detected a new band migrating at around 120 kDa corresponding in size to a putative CB1 dimer. This band was sensitive to reducing agents (50 mM dithiothreitol) and showed sodium dodecylsulphate stability, suggesting the existence of disulfide-linked CB1 dimers in the membrane preparations. Important differences in the anatomical distribution of CB1 were observed with regard to that described previously in monkey and rat; in the human brain, CB1 levels were higher in cortex and caudate than in the cerebellum.


Subject(s)
Brain Chemistry/immunology , Brain/immunology , Cannabinoid Receptor Modulators/metabolism , Receptor, Cannabinoid, CB1/immunology , Age Factors , Antibody Specificity , Brain/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Female , Glycosylation , Humans , Immunohistochemistry/methods , Male , Receptor, Cannabinoid, CB1/metabolism , Subcellular Fractions
16.
Neurochem Int ; 46(3): 213-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15670637

ABSTRACT

Several studies have reported that the chronic administration of opioids induces changes in the biosynthesis of endogenous opioid peptides or their precursors in specific brain regions of the adult central nervous system. However, little is known about the catabolic regulation of opioid peptides and its contribution to neuroadaptative changes underlying drug addiction. In the present study, we have analyzed the activity of two enkephalin-degrading enzymes (puromycin-sensitive aminopeptidase or PSA and aminopeptidase N or APN) and two functionally different, soluble aminopeptidases (aminopeptidase B and aspartyl-aminopeptidase) in postmortem samples of prefrontal cortex and caudate nucleus of eight human heroin addict brains and eight matched-controls. Enzyme activities were fluorimetrically measured using beta-naphthylamide derivatives. An increase in the activity of soluble PSA in the prefrontal cortex of heroin abusers was observed (heroin addict group: 51,452+/-3892 UAP/mg protein versus control group: 42,003+/-2597 UAP/mg protein; P<0.05), while the activity of the other peptidases in both brain regions remained unaltered. This result agrees with previous findings in morphine-tolerant rats, and indicates that soluble PSA may be involved in neurobiological processes which underlie heroin addiction.


Subject(s)
Aminopeptidases/metabolism , Brain/enzymology , Heroin Dependence/enzymology , Adult , CD13 Antigens/metabolism , Caudate Nucleus/metabolism , Endopeptidases/metabolism , Female , Glutamyl Aminopeptidase/metabolism , Humans , Male , Postmortem Changes , Prefrontal Cortex/enzymology , Specimen Handling
18.
J Neurol Neurosurg Psychiatry ; 75(5): 785-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15090584

ABSTRACT

BACKGROUND: Current glioma grading schemes are limited by subjective histological criteria. Imidazoline I(2) receptors are principally expressed on glial cells. OBJECTIVE: To investigate the feasibility of using the measurement of imidazoline I(2) receptor expression to differentiate glial tumours from other types of brain tumours and for grading the different gliomas. METHODS: The specific binding of [(3)H]idazoxan to imidazoline I(2) receptors was measured in homogenates from human gliomas of different grades. RESULTS: The density of imidazoline I(2) receptors was significantly greater in the three types of malignant glial tumours than in postmortem control brain or non-glial tumours. The increase in density correlated with the malignancy grade of the gliomas. No significant differences in affinity values were observed. CONCLUSION: These results suggest that the density of imidazoline I(2) receptors may be a useful radioligand parameter for the differentiation of glial tumours from other types of brain tumours and for grading the different gliomas.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Receptors, Drug/metabolism , Binding Sites , Cell Count , Feasibility Studies , Glial Fibrillary Acidic Protein/metabolism , Glioma/classification , Humans , Idazoxan/metabolism , Imidazoline Receptors , Neoplasm Staging
19.
Mol Psychiatry ; 7(7): 755-67, 2002.
Article in English | MEDLINE | ID: mdl-12192620

ABSTRACT

Abnormalities in the density of neuroreceptors that regulate norepinephrine and serotonin release have been repeatedly reported in brains of suicide victims with mood disorders. Recently, the modulation of the [(35)S]GTPgammaS binding to G-proteins has been introduced as a suitable measure of receptor activity in postmortem human brain. The present study sought to evaluate the function of several G-protein coupled receptors in postmortem brain of suicide victims with mood disorders. Concentration-response curves of the [(35)S]GTPgammaS binding stimulation by selective agonists of alpha(2)-adrenoceptors, 5-HT(1A) serotonin, mu-opioid, GABA(B), and cholinergic muscarinic receptors were performed in frontal cortical membranes from 28 suicide victims with major depression or bipolar disorder and 28 subjects who were matched for gender, age and postmortem delay. The receptor-independent [(35)S]GTPgammaS binding stimulation by mastoparan and the G-protein density were also examined. The alpha(2A)-adrenoceptor-mediated stimulation of [(35)S]GTPgammaS binding with the agonist UK14304 displayed a 4.6-fold greater sensitivity in suicide victims than in controls, without changes in the maximal stimulation. No significant differences were found in parameters of 5-HT(1A) serotonin receptor and other receptor-mediated [(35)S]GTPgammaS binding stimulations. The receptor-independent activation of G-proteins was similar in both groups. Immunoreactive densities of G(alphai1/2)-, G(alphai3)-, G(alphao)-, and G(alphas)-proteins did not differ between suicide victims and controls. In conclusion, alpha(2A)-adrenoceptor sensitivity is increased in the frontal cortex of suicide victims with mood disorders. This receptor supersensitivity is not related to an increased amount or enhanced intrinsic activity of G-proteins. The new finding provides functional support to the involvement of alpha(2)-adrenoceptors in the pathogenesis of mood disorders.


Subject(s)
Brain/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Mood Disorders/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Suicide , Adult , Aged , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Male , Middle Aged , Mood Disorders/diagnosis , Mood Disorders/drug therapy , Psychotropic Drugs/therapeutic use , Radioligand Assay , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT1 , Sulfur Radioisotopes , Tubulin/metabolism
20.
Br J Pharmacol ; 133(8): 1362-70, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11498523

ABSTRACT

1. The effects of desipramine (3 mg kg(-1) i.p.) and clorgyline (1 mg kg(-1) i.p.) on extracellular noradrenaline (NA) in the locus coeruleus (LC) and cingulate cortex were assessed in freely-moving rats by dual-probe microdialysis. Functional activities of alpha(2)-adrenoceptors regulating NA release in the LC and cingulate cortex were determined by systemic (0.3 mg kg(-1) i.p.) or local (0.1 - 100 microM) clonidine administration. 2. Extracellular NA was increased in the LC and cingulate cortex following acute desipramine but not clorgyline treatment. Systemic clonidine decreased NA similarly in desipramine-, clorgyline-, and saline-treated animals, in both brain areas. 3. Long-term (twice daily, 14 days) but not short-term (twice daily, 7 days) desipramine, and long-term clorgyline (once daily, 21 days) treatments increased NA (3 fold) in cingulate cortex but not in the LC. Following long-term treatments, responses of NA to systemic clonidine were attenuated in the LC and cingulate cortex. 4. Clonidine perfusion by reverse dialysis into the cingulate cortex decreased local NA (-55 +/- 9%). The effect was attenuated by long-term desipramine (-31 +/- 9%) and clorgyline (-10 +/- 2%) treatments. 5. Clonidine perfusion by reverse dialysis into the LC decreased NA in the LC (-89 +/- 2%) and in cingulate cortex (-52 +/- 12%). This effect was attenuated in the LC following long-term desipramine (-72 +/- 4%) and clorgyline (-62 +/- 12%) treatments but it was not modified in the cingulate cortex (-57 +/- 10% and -68 +/- 6%, respectively). 6. These findings demonstrate that chronic desipramine or clorgyline treatments increase NA in noradrenergic terminal areas and desensitize alpha(2)-adrenoceptors modulating local NA release at somatodendritic and terminal levels. However, somatodendritic alpha(2)-adrenoceptors that control LC firing activity are not desensitized.


Subject(s)
Brain/drug effects , Brain/metabolism , Clorgyline/pharmacology , Desipramine/pharmacology , Microdialysis , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-Agonists/metabolism , Adrenergic alpha-Agonists/pharmacology , Animals , Clonidine/pharmacology , Clorgyline/administration & dosage , Desipramine/administration & dosage , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...