Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Transl Psychiatry ; 13(1): 384, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38092757

RESUMO

The PD-DLB psychosis complex found in Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB) includes hallucinations, Somatic Symptom/Functional Disorders, and delusions. These disorders exhibit similar presentation patterns and progression. Mechanisms at the root of these symptoms also share similarities with processes promoting altered states of consciousness found in Rapid Eye Movement sleep, psychiatric disorders, or the intake of psychedelic compounds. We propose that these mechanisms find a crucial driver and trigger in the dysregulated activity of high-order thalamic nuclei set in motion by ThalamoCortical Dysrhythmia (TCD). TCD generates the loss of finely tuned cortico-cortical modulations promoted by the thalamus and unleashes the aberrant activity of the Default Mode Network (DMN). TCD moves in parallel with altered thalamic filtering of external and internal information. The process produces an input overload to the cortex, thereby exacerbating DMN decoupling from task-positive networks. These phenomena alter the brain metastability, creating dreamlike, dissociative, or altered states of consciousness. In support of this hypothesis, mind-altering psychedelic drugs also modulate thalamic-cortical pathways. Understanding the pathophysiological background of these conditions provides a conceptual bridge between neurology and psychiatry, thereby helping to generate a promising and converging area of investigation and therapeutic efforts.


Assuntos
Alucinógenos , Doença por Corpos de Lewy , Doenças Neurodegenerativas , Doença de Parkinson , Transtornos Psicóticos , Humanos , Alucinógenos/farmacologia , Doença por Corpos de Lewy/tratamento farmacológico , Doença por Corpos de Lewy/diagnóstico , Doença por Corpos de Lewy/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Tálamo , Doença de Parkinson/tratamento farmacológico
2.
J Psychopharmacol ; 35(4): 469-482, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33645311

RESUMO

BACKGROUND: The reticular thalamus gates thalamocortical information flow via finely tuned inhibition of thalamocortical cells in the mediodorsal thalamus. Brain imaging studies in humans show that the psychedelic lysergic acid diethylamide (LSD) modulates activity and connectivity within the cortico-striato-thalamo-cortical (CSTC) circuit, altering consciousness. However, the electrophysiological effects of LSD on the neurons in these brain areas remain elusive. METHODS: We employed in vivo extracellular single-unit recordings in anesthetized adult male mice to investigate the dose-response effects of cumulative LSD doses (5-160 µg/kg, intraperitoneal) upon reticular thalamus GABAergic neurons, thalamocortical relay neurons of the mediodorsal thalamus, and pyramidal neurons of the infralimbic prefrontal cortex. RESULTS: LSD decreased spontaneous firing and burst-firing activity in 50% of the recorded reticular thalamus neurons in a dose-response fashion starting at 10 µg/kg. Another population of neurons (50%) increased firing and burst-firing activity starting at 40 µg/kg. This modulation was accompanied by an increase in firing and burst-firing activity of thalamocortical neurons in the mediodorsal thalamus. On the contrary, LSD excited infralimbic prefrontal cortex pyramidal neurons only at the highest dose tested (160 µg/kg). The dopamine D2 receptor (D2) antagonist haloperidol administered after LSD increased burst-firing activity in the reticular thalamus neurons inhibited by LSD, decreased firing and burst-firing activity in the mediodorsal thalamus, and showed a trend towards further increasing the firing activity of neurons of the infralimbic prefrontal cortex. CONCLUSION: LSD modulates firing and burst-firing activity of reticular thalamus neurons and disinhibits mediodorsal thalamus relay neurons at least partially in a D2-mediated fashion. These effects of LSD on thalamocortical gating could explain its consciousness-altering effects in humans.


Assuntos
Relação Dose-Resposta a Droga , Fenômenos Eletrofisiológicos , Dietilamida do Ácido Lisérgico/farmacologia , Córtex Pré-Frontal , Tálamo , Animais , Transtornos da Consciência/induzido quimicamente , Transtornos da Consciência/metabolismo , Antagonistas dos Receptores de Dopamina D2/farmacologia , Alucinógenos/farmacologia , Masculino , Camundongos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D2/metabolismo , Tálamo/efeitos dos fármacos , Tálamo/metabolismo
3.
Int J Mol Sci ; 20(10)2019 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-31108968

RESUMO

Melatonin (MLT) is a neurohormone that regulates many physiological functions including sleep, pain, thermoregulation, and circadian rhythms. MLT acts mainly through two G-protein-coupled receptors named MT1 and MT2, but also through an MLT type-3 receptor (MT3). However, the role of MLT receptor subtypes in thermoregulation is still unknown. We have thus investigated the effects of selective and non-selective MLT receptor agonists/antagonists on body temperature (Tb) in rats across the 12/12-h light-dark cycle. Rectal temperature was measured every 15 min from 4:00 a.m. to 9:30 a.m. and from 4:00 p.m. to 9:30 p.m., following subcutaneous injection of each compound at either 5:00 a.m. or 5:00 p.m. MLT (40 mg/kg) had no effect when injected at 5 a.m., whereas it decreased Tb during the light phase only when injected at 5:00 p.m. This effect was blocked by the selective MT2 receptor antagonist 4P-PDOT and the non-selective MT1/MT2 receptor antagonist, luzindole, but not by the α1/MT3 receptors antagonist prazosin. However, unlike MLT, neither the selective MT1 receptor partial agonist UCM871 (14 mg/kg) nor the selective MT2 partial agonist UCM924 (40 mg/kg) altered Tb during the light phase. In contrast, UCM871 injected at 5:00 p.m. increased Tb at the beginning of the dark phase, whereas UCM924 injected at 5:00 a.m. decreased Tb at the end of the dark phase. These effects were blocked by luzindole and 4P-PDOT, respectively. The MT3 receptor agonist GR135531 (10 mg/kg) did not affect Tb. These data suggest that the simultaneous activation of both MT1 and MT2 receptors is necessary to regulate Tb during the light phase, whereas in a complex but yet unknown manner, they regulate Tb differently during the dark phase. Overall, MT1 and MT2 receptors display complementary but also distinct roles in modulating circadian fluctuations of Tb.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Melatonina/administração & dosagem , Receptor MT1 de Melatonina/agonistas , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/agonistas , Acetamidas/administração & dosagem , Acetamidas/farmacologia , Compostos de Anilina/administração & dosagem , Compostos de Anilina/farmacologia , Animais , Injeções Subcutâneas , Masculino , Melatonina/farmacologia , Fotoperíodo , Ratos , Ratos Wistar , Receptor MT1 de Melatonina/antagonistas & inibidores , Receptor MT2 de Melatonina/antagonistas & inibidores , Receptor MT2 de Melatonina/metabolismo , Tetra-Hidronaftalenos/administração & dosagem , Tetra-Hidronaftalenos/farmacologia , Triptaminas/administração & dosagem , Triptaminas/farmacologia
4.
Pain ; 160(1): 136-150, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30157131

RESUMO

Clinical studies indicate that cannabidiol (CBD), the primary nonaddictive component of cannabis that interacts with the serotonin (5-HT)1A receptor, may possess analgesic and anxiolytic effects. However, its effects on 5-HT neuronal activity, as well as its impact on models of neuropathic pain are unknown. First, using in vivo single-unit extracellular recordings in rats, we demonstrated that acute intravenous (i.v.) increasing doses of CBD (0.1-1.0 mg/kg) decreased the firing rate of 5-HT neurons in the dorsal raphe nucleus, which was prevented by administration of the 5-HT1A antagonist WAY 100635 (0.3 mg/kg, i.v.) and the TRPV1 antagonist capsazepine (1 mg/kg, i.v.) but not by the CB1 receptor antagonist AM 251 (1 mg/kg, i.v.). Repeated treatment with CBD (5 mg/kg/day, subcutaneously [s.c.], for 7 days) increased 5-HT firing through desensitization of 5-HT1A receptors. Rats subjected to the spared nerve injury model for 24 days showed decreased 5-HT firing activity, mechanical allodynia, and increased anxiety-like behavior in the elevated plus maze test, open-field test, and novelty-suppressed feeding test. Seven days of treatment with CBD reduced mechanical allodynia, decreased anxiety-like behavior, and normalized 5-HT activity. Antiallodynic effects of CBD were fully prevented by capsazepine (10 mg/kg/day, s.c., for 7 days) and partially prevented by WAY 100635 (2 mg/kg/day, s.c., for 7 days), whereas the anxiolytic effect was blocked only by WAY. Overall, repeated treatment with low-dose CBD induces analgesia predominantly through TRPV1 activation, reduces anxiety through 5-HT1A receptor activation, and rescues impaired 5-HT neurotransmission under neuropathic pain conditions.


Assuntos
Ansiedade/tratamento farmacológico , Ansiedade/etiologia , Canabidiol/uso terapêutico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Neuralgia/complicações , Serotonina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Gânglios Espinais/citologia , Hiperalgesia/terapia , Dietilamida do Ácido Lisérgico/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Neuralgia/patologia , Piperazinas/uso terapêutico , Piperidinas/farmacologia , Pirazóis/farmacologia , Piridinas/uso terapêutico , Ratos , Ratos Wistar , Antagonistas da Serotonina/farmacologia , Natação
5.
Int J Mol Sci ; 17(11)2016 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-27886063

RESUMO

d-Lysergic Acid Diethylamide (LSD) is known for its hallucinogenic properties and psychotic-like symptoms, especially at high doses. It is indeed used as a pharmacological model of psychosis in preclinical research. The goal of this review was to understand the mechanism of action of psychotic-like effects of LSD. We searched Pubmed, Web of Science, Scopus, Google Scholar and articles' reference lists for preclinical studies regarding the mechanism of action involved in the psychotic-like effects induced by LSD. LSD's mechanism of action is pleiotropic, primarily mediated by the serotonergic system in the Dorsal Raphe, binding the 5-HT2A receptor as a partial agonist and 5-HT1A as an agonist. LSD also modulates the Ventral Tegmental Area, at higher doses, by stimulating dopamine D2, Trace Amine Associate receptor 1 (TAAR1) and 5-HT2A. More studies clarifying the mechanism of action of the psychotic-like symptoms or psychosis induced by LSD in humans are needed. LSD's effects are mediated by a pleiotropic mechanism involving serotonergic, dopaminergic, and glutamatergic neurotransmission. Thus, the LSD-induced psychosis is a useful model to test the therapeutic efficacy of potential novel antipsychotic drugs, particularly drugs with dual serotonergic and dopaminergic (DA) mechanism or acting on TAAR1 receptors.


Assuntos
Núcleo Dorsal da Rafe/metabolismo , Alucinógenos/farmacologia , Dietilamida do Ácido Lisérgico/farmacologia , Transtornos Psicóticos/metabolismo , Agonistas do Receptor de Serotonina/farmacologia , Área Tegmentar Ventral/metabolismo , Animais , Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Dopamina/metabolismo , Dopamina/farmacologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/fisiopatologia , Avaliação Pré-Clínica de Medicamentos , Alucinógenos/metabolismo , Humanos , Dietilamida do Ácido Lisérgico/metabolismo , Transtornos Psicóticos/tratamento farmacológico , Transtornos Psicóticos/fisiopatologia , Ratos , Receptor 5-HT1A de Serotonina/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glutamato/metabolismo , Agonistas do Receptor de Serotonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/fisiopatologia
6.
J Pineal Res ; 58(4): 397-417, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25726952

RESUMO

The involvement of melatonin in mammalian brain pathophysiology has received growing interest, but information about the anatomical distribution of its two G-protein-coupled receptors, MT1 and MT2 , remains elusive. In this study, using specific antibodies, we examined the precise distribution of both melatonin receptors immunoreactivity across the adult rat brain using light, confocal, and electron microscopy. Our results demonstrate a selective MT1 and MT2 localization on neuronal cell bodies and dendrites in numerous regions of the rat telencephalon, diencephalon, and mesencephalon. Confocal and ultrastructural examination confirmed the somatodendritic nature of MT1 and MT2 receptors, both being localized on neuronal membranes. Overall, striking differences were observed in the anatomical distribution pattern of MT1 and MT2 proteins, and the labeling often appeared complementary in regions displaying both receptors. Somadendrites labeled for MT1 were observed for instance in the retrosplenial cortex, the dentate gyrus of the hippocampus, the islands of Calleja, the medial habenula, the suprachiasmatic nucleus, the superior colliculus, the substantia nigra pars compacta, the dorsal raphe nucleus, and the pars tuberalis of the pituitary gland. Somadendrites endowed with MT2 receptors were mostly observed in the CA3 field of the hippocampus, the reticular thalamic nucleus, the supraoptic nucleus, the inferior colliculus, the substantia nigra pars reticulata, and the ventrolateral periaqueductal gray. Together, these data provide the first detailed neurocytological mapping of melatonin receptors in the adult rat brain, an essential prerequisite for a better understanding of melatonin distinct receptor function and neurophysiology.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/metabolismo , Animais , Western Blotting , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley
7.
Pain ; 156(2): 305-317, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25599452

RESUMO

Neuropathic pain is an important public health problem for which only a few treatments are available. Preclinical studies show that melatonin (MLT), a neurohormone acting on MT1 and MT2 receptors, has analgesic properties, likely through MT2 receptors. Here, we determined the effects of the novel selective MLT MT2 receptor partial agonist N-{2-([3-bromophenyl]-4-fluorophenylamino)ethyl}acetamide (UCM924) in 2 neuropathic pain models in rats and examined its supraspinal mechanism of action. In rat L5-L6 spinal nerve ligation and spared nerve injury models, UCM924 (20-40 mg/kg, subcutaneously) produced a prolonged antinociceptive effect that is : (1) dose-dependent and blocked by the selective MT2 receptor antagonist 4-phenyl-2-propionamidotetralin, (2) superior to a high dose of MLT (150 mg/kg) and comparable with gabapentin (100 mg/kg), but (3) without noticeable motor coordination impairments in the rotarod test. Using double staining immunohistochemistry, we found that MT2 receptors are expressed by glutamatergic neurons in the rostral ventrolateral periaqueductal gray. Using in vivo electrophysiology combined with tail flick, we observed that microinjection of UCM924 into the ventrolateral periaqueductal gray decreased tail flick responses, depressed the firing activity of ON cells, and activated the firing of OFF cells; all effects were MT2 receptor-dependent. Altogether, these data demonstrate that selective MT2 receptor partial agonists have analgesic properties through modulation of brainstem descending antinociceptive pathways, and MT2 receptors may represent a novel target in the treatment of neuropathic pain.


Assuntos
Acetamidas/metabolismo , Compostos de Anilina/metabolismo , Tronco Encefálico/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Tratos Piramidais/metabolismo , Receptor MT2 de Melatonina/metabolismo , Acetamidas/farmacologia , Acetamidas/uso terapêutico , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Animais , Tronco Encefálico/efeitos dos fármacos , Ligantes , Masculino , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Tratos Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Receptor MT2 de Melatonina/agonistas
8.
J Neurosci ; 31(50): 18439-52, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-22171046

RESUMO

Melatonin activates two brain G-protein coupled receptors, MT(1) and MT(2), whose differential roles in the sleep-wake cycle remain to be defined. The novel MT(2) receptor partial agonist, N-{2-[(3-methoxyphenyl) phenylamino] ethyl} acetamide (UCM765), is here shown to selectively promote non-rapid eye movement sleep (NREMS) in rats and mice. The enhancement of NREMS by UCM765 is nullified by the pharmacological blockade or genetic deletion of MT(2) receptors. MT(2), but not MT(1), knock-out mice show a decrease in NREMS compared to the wild strain. Immunohistochemical labeling reveals that MT(2) receptors are localized in sleep-related brain regions, and notably the reticular thalamic nucleus (Rt). Microinfusion of UCM765 in the Rt promotes NREMS, and its systemic administration induces an increase in firing and rhythmic burst activity of Rt neurons, which is blocked by the MT(2) antagonist 4-phenyl-2-propionamidotetralin. Since developing hypnotics that increase NREMS without altering sleep architecture remains a medical challenge, MT(2) receptors may represent a novel target for the treatment of sleep disorders.


Assuntos
Acetamidas/farmacologia , Compostos de Anilina/farmacologia , Neurônios/efeitos dos fármacos , Receptor MT2 de Melatonina/metabolismo , Sono/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Receptor MT2 de Melatonina/agonistas , Receptor MT2 de Melatonina/genética
9.
Psychopharmacology (Berl) ; 214(2): 465-76, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21042794

RESUMO

RATIONALE: By enhancing brain anandamide tone, inhibitors of fatty acid amide hydrolase (FAAH) induce anxiolytic-like effects in rodents and enhance brain serotonergic transmission. Mice lacking the faah gene (FAAH(-/-)) show higher anandamide levels. However, their emotional phenotype is still debated and their brain serotonergic tone remained unexplored. OBJECTIVES AND METHODS: In this study, we tested FAAH(-/-) mice in the social interaction and the open field tests performed under different lighting conditions (dim and bright) since variations of the experimental context were proposed to explain opposite findings. Moreover, by microdialysis performed under dim light, we analyzed their serotonergic transmission in frontal cortex (FC) and ventral hippocampus (vHIPP). RESULTS: In both light conditions, FAAH(-/-) mice showed reduced emotionality, compared to wt controls, as suggested by the increased rearing and reduced thigmotaxis displayed in the open field and by the longer time spent in social interaction. Basal serotonergic tone was higher in the FC of mutant mice as compared to control mice, while no difference was observed in the vHIPP. K(+)-induced depolarization produced similar increases of serotonin in both areas of both genotypes. An acute treatment with the CB1 antagonist rimonabant completely abolished the emotional phenotype of FAAH(-/-) mice and prevented the K(+)-stimulated release of serotonin in their FC and vHIPP, without producing any effect in wt mice. CONCLUSIONS: Our results support the role of FAAH in the regulation of emotional reactivity and suggest that anandamide-mediated hyperactivation of CB1 is responsible for the emotional phenotype of FAAH(-/-) mice and for their enhanced serotonergic tone.


Assuntos
Amidoidrolases/deficiência , Comportamento Animal , Emoções , Lobo Frontal/metabolismo , Hipocampo/metabolismo , Serotonina/metabolismo , Transmissão Sináptica , Amidoidrolases/genética , Análise de Variância , Animais , Ácidos Araquidônicos/metabolismo , Comportamento Animal/efeitos dos fármacos , Distribuição de Qui-Quadrado , Emoções/efeitos dos fármacos , Endocanabinoides , Lobo Frontal/citologia , Lobo Frontal/efeitos dos fármacos , Genótipo , Habituação Psicofisiológica , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdiálise , Atividade Motora , Neurônios/metabolismo , Fenótipo , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/metabolismo , Potássio/metabolismo , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Rimonabanto , Comportamento Social , Transmissão Sináptica/efeitos dos fármacos , Fatores de Tempo
10.
Neurobiol Dis ; 37(3): 641-55, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19969082

RESUMO

The pathophysiological neural mechanism underlying the depressogenic and anxiogenic effects of chronic adolescent cannabinoid use may be linked to perturbations in monoaminergic neurotransmission. We tested this hypothesis by administering the CB(1) receptor agonist WIN55,212-2, once daily for 20 days to adolescent and adult rats, subsequently subjecting them to tests for emotional reactivity paralleled by the in vivo extracellular recordings of serotonergic and noradrenergic neurons. Chronic adolescent exposure but not adult exposure to low (0.2 mg/kg) and high (1.0 mg/kg) doses led to depression-like behaviour in the forced swim and sucrose preference test, while the high dose also induced anxiety-like consequences in the novelty-suppressed feeding test. Electrophysiological recordings revealed both doses to have attenuated serotonergic activity, while the high dose also led to a hyperactivity of noradrenergic neurons only after adolescent exposure. These suggest that long-term exposure to cannabinoids during adolescence induces anxiety-like and depression-like behaviours in adulthood and that this may be instigated by serotonergic hypoactivity and noradrenergic hyperactivity.


Assuntos
Monoaminas Biogênicas/agonistas , Monoaminas Biogênicas/antagonistas & inibidores , Encéfalo/efeitos dos fármacos , Canabinoides/toxicidade , Abuso de Maconha/fisiopatologia , Transtornos Mentais/induzido quimicamente , Fatores Etários , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Transtornos de Ansiedade/induzido quimicamente , Transtornos de Ansiedade/metabolismo , Transtornos de Ansiedade/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Benzoxazinas/toxicidade , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Química Encefálica/efeitos dos fármacos , Química Encefálica/fisiologia , Bloqueadores dos Canais de Cálcio/toxicidade , Transtorno Depressivo/induzido quimicamente , Transtorno Depressivo/metabolismo , Transtorno Depressivo/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Emoções/efeitos dos fármacos , Emoções/fisiologia , Masculino , Abuso de Maconha/metabolismo , Abuso de Maconha/psicologia , Transtornos Mentais/metabolismo , Morfolinas/toxicidade , Naftalenos/toxicidade , Norepinefrina/metabolismo , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Tempo
11.
Trends Pharmacol Sci ; 30(9): 484-93, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19732971

RESUMO

The endocannabinoid system is a neuromodulatory system which is known to regulate emotional, cognitive, neurovegetative and motivational processes. Substantial evidence has accumulated implicating a deficit in endocannabinoid in the etiology of depression; accordingly, pharmacological augmentation of endocannabinoid signaling could be a novel target for the pharmacotherapy of depression. Within preclinical models, facilitation of endocannabinoid neurotransmission evokes both antidepressant and anxiolytic effects. Similar to the actions of conventional antidepressants, enhancement of endocannabinoid signaling can enhance serotonergic and noradrenergic transmission; increase cellular plasticity and neurotrophin expression within the hippocampus; and dampen activity within the neuroendocrine stress axis. Furthermore, limbic endocannabinoid activity is increased by both pharmacological and somatic treatments for depression, and, in turn, appears to contribute to some of the neuroadaptive alterations elicited by these treatments. These preclinical findings support the rationale for the clinical development of agents which inhibit the cellular uptake and/or metabolism of endocannabinoids in the treatment of mood disorders.


Assuntos
Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Animais , Moduladores de Receptores de Canabinoides/metabolismo , Ensaios Clínicos como Assunto , Depressão/fisiopatologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Endocanabinoides , Humanos , Transtornos do Humor/tratamento farmacológico , Transtornos do Humor/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
12.
Curr Pharm Des ; 15(14): 1623-46, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19442178

RESUMO

Among all mental disorders, major depression has the highest rate of prevalence and incidence of morbidity. Currently available antidepressant therapies have limited efficacies; consequently, research on new drugs for the treatment of mood disorders has become increasingly critical. Recent preclinical evidences that cannabinoid agonists and endocannabinoid enhancers, such as the fatty acid amide hydrolase (FAAH) inhibitors, can impact mood regulation have opened a new line of research in antidepressant drug discovery. However, the neurobiological mechanisms linking the endocannabinoid system with the pathophysiology of mood disorders and antidepressant action remain unclarified. In this review, we have presented an update on preclinical data indicating the antidepressant potential of cannabinoid agonists and endocannabinoid enhancers in comparison to standard antidepressants. Data obtained from CB(1) knockout (CB(1)-/-) and FAAH knockout (FAAH-/-) mice have also been examined within this context. We have illustrated how the various classes of antidepressants exert their therapeutic action. In particular, all antidepressants increase the neurotransmission of serotonin after long-term treatment, enhance the tonic activity of hippocampal 5-HT(1A) receptors, promote neurogenesis, and modulate (decrease or increase) the firing activity of noradrenergic neurons. Interestingly, cannabinoid agonists and endocannabinoid enhancers increase serotonin and noradrenergic neuronal firing activity, increase serotonin release in the hippocampus, as well as promote neurogenesis. Since cannabinoid-derived drugs potentiate monoaminergic neurotransmission and hippocampal neurogenesis through distinct pathways compared to classical antidepressants, they may represent an alternative drug class in the pharmacotherapy of mood and other neuropsychiatric disorders.


Assuntos
Antidepressivos/farmacologia , Canabinoides/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Animais , Moduladores de Receptores de Canabinoides/metabolismo , Transtorno Depressivo Maior/fisiopatologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Endocanabinoides , Humanos , Camundongos , Camundongos Knockout , Neurogênese/efeitos dos fármacos , Ratos , Receptor CB1 de Canabinoide/agonistas , Serotonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA