Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34083436

RESUMO

Prefrontal control of cognitive functions critically depends upon glutamatergic transmission and N-methyl D-aspartate (NMDA) receptors, the activity of which is regulated by dopamine. Yet whether the NMDA receptor coagonist d-serine is implicated in the dopamine-glutamate dialogue in the prefrontal cortex (PFC) and other brain areas remains unexplored. Here, using electrophysiological recordings, we show that d-serine is required for the fine-tuning of glutamatergic neurotransmission, neuronal excitability, and synaptic plasticity in the PFC through the actions of dopamine at D1 and D3 receptors. Using in vivo microdialysis, we show that D1 and D3 receptors exert a respective facilitatory and inhibitory influence on extracellular levels and activity of d-serine in the PFC, with actions expressed primarily via the cAMP/protein kinase A (PKA) signaling cascade. Further, using functional magnetic resonance imaging (fMRI) and behavioral assessment, we show that d-serine is required for the potentiation of cognition by D3R blockade as revealed in a test of novel object recognition memory. Collectively, these results unveil a key role for d-serine in the dopaminergic neuromodulation of glutamatergic transmission and PFC activity, findings with clear relevance to the pathogenesis and treatment of diverse brain disorders involving alterations in dopamine-glutamate cross-talk.


Assuntos
Dopamina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Animais , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Camundongos Knockout , Racemases e Epimerases/deficiência , Racemases e Epimerases/genética , Receptores Dopaminérgicos/metabolismo , Esquizofrenia , Transmissão Sináptica/efeitos dos fármacos
2.
Neural Plast ; 2018: 4196961, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29531524

RESUMO

The antiparkinsonian ropinirole and pramipexole are D3 receptor- (D3R-) preferring dopaminergic (DA) agonists used as adjunctive therapeutics for the treatment resistant depression (TRD). While the exact antidepressant mechanism of action remains uncertain, a role for D3R in the restoration of impaired neuroplasticity occurring in TRD has been proposed. Since D3R agonists are highly expressed on DA neurons in humans, we studied the effect of ropinirole and pramipexole on structural plasticity using a translational model of human-inducible pluripotent stem cells (hiPSCs). Two hiPSC clones from healthy donors were differentiated into midbrain DA neurons. Ropinirole and pramipexole produced dose-dependent increases of dendritic arborization and soma size after 3 days of culture, effects antagonized by the selective D3R antagonists SB277011-A and S33084 and by the mTOR pathway kinase inhibitors LY294002 and rapamycin. All treatments were also effective in attenuating the D3R-dependent increase of p70S6-kinase phosphorylation. Immunoneutralisation of BDNF, inhibition of TrkB receptors, and blockade of MEK-ERK signaling likewise prevented ropinirole-induced structural plasticity, suggesting a critical interaction between BDNF and D3R signaling pathways. The highly similar profiles of data acquired with DA neurons derived from two hiPSC clones underpin their reliability for characterization of pharmacological agents acting via dopaminergic mechanisms.


Assuntos
Antiparkinsonianos/administração & dosagem , Benzotiazóis/administração & dosagem , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios Dopaminérgicos , Indóis/administração & dosagem , Plasticidade Neuronal/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Pramipexol , Transdução de Sinais
3.
Mol Pharmacol ; 83(6): 1176-89, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23543412

RESUMO

Although long-term exposure to nicotine is highly addictive, one beneficial consequence of chronic tobacco use is a reduced risk for Parkinson's disease. Of interest, these effects both reflect structural and functional plasticity of brain circuits controlling reward and motor behavior and, specifically, recruitment of nicotinic acetylcholine receptors (nAChR) in mesencephalic dopaminergic neurons. Because the underlying cellular mechanisms are poorly understood, we addressed this issue with use of primary cultures of mouse mesencephalic dopaminergic neurons. Exposure to nicotine (1-10 µM) for 72 hours in vitro increased dendritic arborization and soma size in primary cultures. These effects were blocked by mecamylamine and dihydro-ß-erythroidine, but not methyllycaconitine. The involvement of α4ß2 nAChR was supported by the lack of nicotine-induced structural remodeling in neurons from α4 null mutant mice (KO). Challenge with nicotine triggered phosphorylation of the extracellular signal-regulated kinase (ERK) and the thymoma viral proto-oncogene (Akt), followed by activation of the mammalian target of rapamycin complex 1 (mTORC1)-dependent p70 ribosomal S6 protein kinase. Upstream pathway blockade using the phosphatidylinositol 3-kinase inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one hydrochloride] resulted in suppression of nicotine-induced phosphorylations and structural plasticity. These effects were dependent on functional DA D3 receptor (D3R), because nicotine was inactive both in cultures from D3R KO mice and after pharmacologic blockade with D3R antagonist trans-N-4-2-(6-cyano-1,2,3, 4-tetrahydroisoquinolin-2-yl)ethylcyclohexyl-4-quinolinecarboxamide (SB-277011-A) (50 nM). Finally, exposure to nicotine in utero (5 mg/kg/day for 5 days) resulted in increased soma area of DAergic neurons of newborn mice, effects not observed in D3 receptor null mutant mice mice. These findings indicate that nicotine-induced structural plasticity at mesencephalic dopaminergic neurons involves α4ß2 nAChRs together with dopamine D3R-mediated recruitment of ERK/Akt-mTORC1 signaling.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Mesencéfalo/efeitos dos fármacos , Nicotina/farmacologia , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Dopamina D3/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Ativação Enzimática , Feminino , Sistema de Sinalização das MAP Quinases , Exposição Materna , Troca Materno-Fetal , Alvo Mecanístico do Complexo 1 de Rapamicina , Mesencéfalo/citologia , Mesencéfalo/metabolismo , Camundongos , Camundongos Knockout , Complexos Multiproteicos , Gravidez , Receptores de Dopamina D3/genética , Receptores Nicotínicos/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR
4.
Curr Top Behav Neurosci ; 60: 73-87, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35538302

RESUMO

Structural neuroplasticity in the adult brain is a process involving quantitative changes of the number and size of neurons and of their dendritic arborization, axon branching, spines, and synapses. These changes can occur in specific neural circuits as adaptive response to environmental challenges, exposure to stressors, tissue damage or degeneration. Converging studies point to evidence of structural plasticity in circuits operated by glutamate, GABA, dopamine, and serotonin neurotransmitters, in concert with neurotrophic factors such as Brain Derived Neurotrophic Factor (BDNF) or Insulin Growth Factor 1 (IGF1) and a series of modulators that include circulating hormones. Intriguingly, most of these endogenous agents trigger the activation of the PI3K/Akt/mTOR and ERK1/2 intracellular pathways that, in turn, lead to the production of growth-related structural changes, enhancing protein synthesis, metabolic enzyme functions, mitogenesis for energy, and new lipid-bilayer membrane apposition. The dopamine (DA) D3 receptor has been shown to play a specific role by inducing structural plasticity of the DAergic neurons of the nigrostriatal and mesocorticolimbic circuit, where they are expressed in rodents and humans, via activation of the mTORC1 and ERK1/2 pathways. These effects are BDNF-dependent and require the recruitment of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors to allow the structural changes. Since in mood disorders, depression and anhedonia have been proposed to be associated with impaired neuroplasticity and reduced DAergic tone in brain circuits connecting prefrontal cortex, ventral striatum, amygdala, and ventral mesencephalon, activation of D3 receptors could provide a therapeutic benefit. Sustained improvements of mood and anhedonia were observed in subjects with an unsatisfactory response to serotonin uptake inhibitors (SSRI) when treated with D3-preferential D2/D3 agonists such as pramipexole and ropinirole. The recent evidence that downstream mTOR pathway activation in human mesencephalic DA neurons is also produced by ketamine, probably the most effective antidepressant currently used in subjects with treatment-resistant depression, further supports the rationale for a D3 receptor activation in mood disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Receptores de Dopamina D3 , Humanos , Receptores de Dopamina D3/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dopamina , Anedonia , Depressão , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia , Encéfalo/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/farmacologia , Neurônios Dopaminérgicos/metabolismo , Plasticidade Neuronal
5.
J Neurochem ; 120(5): 765-78, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22145570

RESUMO

Exposure to psychostimulants results in neuroadaptive changes of the mesencephalic dopaminergic system including morphological reorganization of dopaminergic neurons. Increased dendrite arborization and soma area were previously observed in primary cultures of mesencephalic dopaminergic neurons after 3-day exposure to dopamine agonists via activation of D(3) autoreceptors (D(3) R). In this work, we showed that cocaine significantly increased dendritic arborization and soma area of dopaminergic neurons from E12.5 mouse embryos by activating phosphorylation of extracellular signal-regulated kinase (ERK) and thymoma viral proto-oncogene (Akt). These effects were dependent on functional D(3) R expression because cocaine did not produce morphological changes or ERK/Akt phosphorylation neither in primary cultures of D(3) R mutant mice nor following pharmacologic blockade with D(3) R antagonists SB-277011-A and S-33084. Cocaine effects on morphology and ERK/Akt phosphorylation were inhibited by pre-incubation with the phosphatidylinositol 3-kinase inhibitor LY294002. These observations were corroborated in vivo by morphometrical assessment of mesencephalic dopaminergic neurons of P1 newborns exposed to cocaine from E12.5 to E16.5. Cocaine increased the soma area of wild-type but not of D(3) R mutant mice, supporting the translational value of primary culture. These findings indicate a direct involvement of D3R and ERK/Akt pathways as critical mediators of cocaine-induced structural plasticity, suggesting their involvement in psychostimulant addiction.


Assuntos
Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Mesencéfalo/citologia , Neurônios/fisiologia , Terminações Pré-Sinápticas/metabolismo , Receptores de Dopamina D3/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Dopamina , Dopaminérgicos/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Proteína Oncogênica v-akt/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Receptores de Dopamina D3/deficiência , Transdução de Sinais/efeitos dos fármacos , Trítio/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
Front Pharmacol ; 13: 913210, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721218

RESUMO

The epigenetic agents, L-acetylcarnitine (LAC) and L-methylfolate (MF) are putative candidates as add-on drugs in depression. We evaluated the effect of a combined treatment with LAC and MF in two different paradigms of chronic stress in mice and in human inducible pluripotent stem cells (hiPSCs) differentiated into dopaminergic neurons. Two groups of mice were exposed to chronic unpredictable stress (CUS) for 28 days or chronic restraint stress (CRS) for 21 day, and LAC (30 or 100 mg/kg) and/or MF (0.75 or 3 mg/kg) were administered i.p. once a day for 14 days, starting from the last week of stress. In both stress paradigms, LAC and MF acted synergistically in reducing the immobility time in the forced swim test and enhancing BDNF protein levels in the frontal cortex and hippocampus. In addition, LAC and MF acted synergistically in enhancing type-2 metabotropic glutamate receptor (mGlu2) protein levels in the hippocampus of mice exposed to CRS. Interestingly, CRS mice treated with MF showed an up-regulation of NFκB p65, which is a substrate for LAC-induced acetylation. We could also demonstrate a synergism between LAC and MF in cultured hiPSCs differentiated into dopamine neurons, by measuring dendrite length and number, and area of the cell soma after 3 days of drug exposure. These findings support the combined use of LAC and MF in the treatment of MDD and other stress-related disorders.

7.
J Neurochem ; 117(2): 253-63, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21272002

RESUMO

We report a novel mechanism for dopamine D(1) receptor (D(1) R)-mediated extracellular signal-regulated kinases (Erk) activation in rat striatum. Erk signaling depends on phosphorylation and dephosphorylation events mediated by specific kinases and phosphatases. The tyrosine phosphatase Shp-2, that is required for Erk activation by tyrosine kinase receptors, has been recently shown to regulate signaling downstream of few G protein-coupled receptors. We show that the D(1) R interacts with Shp-2, that D(1) R stimulation results in Shp-2 tyrosine phosphorylation and activation in primary striatal neuronal cultures and that D(1) R/Shp-2 interaction is required for transmitting D(1) R-dependent signaling to Erk1/2 activation. D(1) R-mediated Erk1/2 phosphorylation in cultured striatal neurons is in fact abolished by over-expression of the inactive Shp-2(C/S) mutant and by small interfering RNA-induced Shp-2 silencing. Moreover, by using selective inhibitors we show that both D(1) R-induced Shp-2 activation and Erk1/2 phosphorylation are dependent on the cyclic AMP/protein kinase A pathway and require Src. These results, which were substantiated also in transfected human embryonic kidney 293 cells, provide a novel mechanism by which to converge D(1) R signaling to the Erk pathway and suggest that Shp-2 or the D(1) R/Shp-2 interface could represent a potential drug target for disorders of dopamine transmission involving malfunctioning of D(1) R signaling.


Assuntos
Corpo Estriado/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neurônios/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/fisiologia , Adenilil Ciclases/metabolismo , Análise de Variância , Animais , Anticorpos/farmacologia , Benzazepinas/farmacologia , Células Cultivadas , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Embrião de Mamíferos , Humanos , Imunoprecipitação/métodos , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/imunologia , RNA Interferente Pequeno/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção/métodos , Tirosina/metabolismo
9.
Eur J Pharmacol ; 890: 173635, 2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33065094

RESUMO

F17464 (N-(3-{4-[4-(8-Oxo-8H-[1,3]-dioxolo-[4,5-g]-chromen-7-yl)-butyl]-piperazin-1-yl}-phenyl)-methanesulfonamide, hydrochloride) is a new potential antipsychotic with a unique profile. The compound exhibits high affinity for the human dopamine receptor subtype 3 (hD3) (Ki = 0.17 nM) and the serotonin receptor subtype 1a (5-HT1a) (Ki = 0.16 nM) and a >50 fold lower affinity for the human dopamine receptor subtype 2 short and long form (hD2s/l) (Ki = 8.9 and 12.1 nM, respectively). [14C]F17464 dynamic studies show a slower dissociation rate from hD3 receptor (t1/2 = 110 min) than from hD2s receptor (t1/2 = 1.4 min) and functional studies demonstrate that F17464 is a D3 receptor antagonist, 5-HT1a receptor partial agonist. In human dopaminergic neurons F17464 blocks ketamine induced morphological changes, an effect D3 receptor mediated. In vivo F17464 target engagement of both D2 and 5-HT1a receptors is demonstrated in displacement studies in the mouse brain. F17464 increases dopamine release in the rat prefrontal cortex and mouse lateral forebrain - dorsal striatum and seems to reduce the effect of MK801 on % c-fos mRNA medium expressing neurons in cortical and subcortical regions. F17464 also rescues valproate induced impairment in a rat social interaction model of autism. All the neurochemistry and behavioural effects of F17464 are observed in the dose range 0.32-2.5 mg/kg i.p. in both rats and mice. The in vitro - in vivo pharmacology profile of F17464 in preclinical models is discussed in support of a therapeutic use of the compound in schizophrenia and autism.


Assuntos
Antipsicóticos/farmacologia , Benzopiranos/farmacologia , Antagonistas de Dopamina/farmacologia , Piperazinas/farmacologia , Receptores de Dopamina D3/antagonistas & inibidores , Sulfonamidas/farmacologia , Animais , Antipsicóticos/uso terapêutico , Transtorno Autístico/induzido quimicamente , Transtorno Autístico/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Benzopiranos/uso terapêutico , Monoaminas Biogênicas/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Catalepsia/tratamento farmacológico , Células Cultivadas , Dopamina/metabolismo , Antagonistas de Dopamina/uso terapêutico , Neurônios Dopaminérgicos/efeitos dos fármacos , Feminino , Genes fos/efeitos dos fármacos , Masculino , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Piperazinas/uso terapêutico , Prolactina/sangue , Ratos Sprague-Dawley , Receptores de Dopamina D3/metabolismo , Sulfonamidas/uso terapêutico , Ácido Valproico/toxicidade
10.
J Recept Signal Transduct Res ; 30(5): 347-54, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20684667

RESUMO

CONTEXT: G protein-coupled receptors (GPCRs) have been classically thought to work as monomeric entities. The current view of their organization, however, assumes that they are part of highly organized molecular complexes, where different receptors and interacting proteins are clustered. These heteromers have peculiar pharmacological, signaling, and trafficking properties. GPCR heteromerization, raising different combinatorial possibilities, thus underlies an unexpected level of diversity within this receptor family. METHODS: In this paper, we summarize recent data, reported by different research groups, suggesting that the dopamine (DA) D1 receptor forms heteromers with receptors of the same family and with structurally and functionally divergent receptors. RESULTS AND DISCUSSION: DA D1 and D3 receptors and glutamate NMDA receptors regulate rewarding mechanisms and motivated behavior, modulate emotional and cognitive processes and regulate locomotor activity by extensive cross-talk mechanisms. Co-localization of D1 and D3 receptors and D1 and NMDA receptors in specific neuronal populations in the striatum and nucleus accumbens, moreover, suggested that their cross-talk may involve direct interactions. By using different experimental approaches various groups have, in fact, demonstrated the existence of D1-NMDA and D1-D3 heteromers, in both transfected cell systems and in the straitum, with peculiar pharmacological, signaling, and functional properties. The putative role of the D1-D3 and D1-NMDA heteromers in the physiological regulation of striatal function and in the development of motor dysfunctions will be discussed.


Assuntos
Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Quaternária de Proteína , Receptores Dopaminérgicos/química , Receptores Dopaminérgicos/metabolismo , Animais , Dimerização , Humanos , Transtornos dos Movimentos/metabolismo , Isoformas de Proteínas/genética , Receptores Dopaminérgicos/genética , Transdução de Sinais/fisiologia
11.
Front Neurosci ; 14: 632, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32625059

RESUMO

Negative symptoms (NS) represent a heterogeneous dimension of schizophrenia (SCZ), associated with a poor functional outcome. A dysregulated dopamine (DA) system, including a reduced D1 receptor activation in the prefrontal cortex, DA hypoactivity in the caudate and alterations in D3 receptor activity, seems to contribute to the pathogenesis of NS. However, failure to take into account the NS heterogeneity has slowed down progress in research on their neurobiological correlates and discoveries of new effective treatments. A better neurobiological characterization of NS is needed, and this requires objective quantification of their features that can be applied in translational models, such as animal models and human inducible pluripotent stem cells (iPSC). In this review we summarize the evidence for dopaminergic alterations relevant to NS in translational animal models focusing on dysfunctional motivation, a core aspect of NS. Among others, experiments on mutant rodents with an overexpression of DA D2 or D3 receptors and the dopamine deficient mice are discussed. In the second part we summarize the findings from recent studies using iPSC to model the pathogenesis of SCZ. By retaining the genetic background of risk genetic variants, iPSC offer the possibility to study the effect of de novo mutations or inherited polymorphisms from subgroups of patients and their response to drugs, adding an important tool for personalized psychiatry. Given the key role of DA in NS, we focus on findings of iPSC-derived DA neurons. Since implementation of iPSC-derived neurons to study the neurobiology of SCZ is a relatively recent acquisition, the available data are limited. We highlight some methodological aspects of relevance in the interpretation of in vitro testing results, including limitations and strengths, offering a critical viewpoint for the implementation of future pharmacological studies aimed to the discovery and characterization of novel treatments for NS.

12.
Chronic Stress (Thousand Oaks) ; 3: 2470547019842545, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32440593

RESUMO

The mechanisms underlying the antidepressant effects of ketamine in treatment-resistant depression are only partially understood. Reactivation of neural plasticity in prefrontal cortex has been considered critical in mediating the effects of standard antidepressants, but in treatment-resistant depression patients with severe anhedonia, other components of the affected brain circuits, for example, the dopamine system, could be involved. In a recent article in Molecular Psychiatry, we showed that ketamine induces neural plasticity in human and mouse dopaminergic neurons. Human dopaminergic neurons were differentiated from inducible pluripotent stem cells for over 60 days. Mimicking the pharmacokinetic exposures occurring in treatment-resistant depression subjects, cultures were incubated with either ketamine at 0.1 and 1 µM for 1 h or with its active metabolite (2R,6R)-hydroxynorketamine at 0.1 and 0.5 µM for up to 6 h. Three days after dosing, we observed a concentration-dependent increase in dendritic arborization and soma size. These effects were mediated by the activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor that triggered the pathways of mammalian target of rapamycin and extracellular signal-regulated kinase via the engagement of brain-derived neurotrophic factor signaling, as previously described in rodent prefrontal cortex. Interestingly, we found that neural plasticity induced by ketamine requires functionally intact dopamine D3 receptors. These data are in keeping with our recent observation that plasticity can be induced in human dopaminergic neurons by the D3 receptor-preferential agonist pramipexole, whose effect as augmentation treatment in treatment-resistant depression has been reported. Overall, the evidence of pharmacologic response in human inducible pluripotent stem cell-derived neurons could provide complementary information to those provided by circuit-based imaging when assessing the potential response to a given augmentation treatment.

13.
Neuroreport ; 30(3): 207-212, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30586092

RESUMO

The mechanisms underlying the prolonged antidepressant effects after a single exposure to ketamine are only partially understood. Converging findings indicate a critical role of structural neuroplasticity, recently also proposed for dopaminergic (DA) neurons known to be involved in a depression core symptom, anhedonia. We recently showed that ketamine induces dendritic outgrowth in human DA neurons differentiated in-vitro from induced pluripotent stem cells of healthy donors, a phenomenon blocked by the α-amino-3-hydroxy-5-methy-4-isoxazole propionate receptor antagonist NBQX. As changes in the expression of AMPA receptor subunits GluR1 and GluR2 were observed in neuroplasticity of rodent DA neurons, we aimed to explore this phenomenon in human DA neurons. Using specific antibodies against GluR1 and GluR2 α-amino-3-hydroxy-5-methy-4-isoxazole propionate receptor subunits, we showed that GluR1 levels were significantly higher in soma than in dendrites, whereas for GluR2, levels were significantly higher in dendrites than in soma. One hour exposure to 1 µM ketamine increased the signal of both subunits in dendrites, but only of GluR2 in soma, at 24, 48, and 72 h. Nonlinear polynomial fitting of dendritic expression indicated that the two curves were significantly different, with stronger and more sustained effects on GluR2 expression. Overall, these data support a role for GluR1 and GluR2 dendritic upregulation in driving structural plasticity in human DA neurons depending on ketamine transient exposure, indicating translationally relevant downstream mechanism possibly involved in antidepressant effects.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Ketamina/farmacologia , Dendritos/metabolismo , Neurônios Dopaminérgicos/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Plasticidade Neuronal/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos
14.
Biochem Pharmacol ; 163: 154-168, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30772268

RESUMO

In this paper, we designed, synthesized and tested a small set of three new derivatives potentially targeting the D3R-nAChR heteromer, a receptor complex recently identified and characterized as the molecular entity that, in dopaminergic neurons, mediates the neurotrophic effects of nicotine. By means of a partially rigidified spacer of variable length, we incorporated in the new compounds (1a-c) the pharmacophoric substructure of a known ß2-subunit-containing nAChR agonist (A-84543) and that of the D2/D3R agonist drug ropinirole. All the compounds retained the ability to bind with high affinity both ß2-subunit-containing nAChR and D3R. Compound 1a, renamed HyNDA-1, which is characterized by the shortest linker moiety, was the most interesting ligand. We found, in fact, that HyNDA-1 significantly modulated structural plasticity on both mice and human dopaminergic neurons, an effect strongly prevented by co-incubating this ligand with either nAChR or D3R antagonists. Moreover, the neurotrophic effects of HyNDA-1 were specifically lost by disrupting the complex with specific interfering peptides. Interestingly, by using the Bioluminescence Resonance Energy Transfer 2 (BRET2) assay in HEK-293 transfected cells, we also found that HyNDA-1 has the ability to increase the affinity of interaction between nAChR and D3R. Overall, our results indicate that the neurotrophic effects of HyNDA-1 are mediated by activation of the D3R-nAChR heteromeric complex specifically expressed on dopaminergic neurons.


Assuntos
Agonistas de Dopamina/administração & dosagem , Neurônios Dopaminérgicos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Agonistas Nicotínicos/administração & dosagem , Receptores de Dopamina D3/metabolismo , Receptores Nicotínicos/metabolismo , Animais , Células Cultivadas , Agonistas de Dopamina/química , Neurônios Dopaminérgicos/efeitos dos fármacos , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Agonistas Nicotínicos/química , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D3/agonistas
15.
J Neurochem ; 106(2): 560-77, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18410503

RESUMO

Progressive degeneration and intraneuronal Lewy bodies made of filamentous alpha-synuclein (alpha-syn) in dopaminergic cells of the nigrostriatal system are characteristics of Parkinson's disease (PD). Glucose uptake is reduced in some of the brain regions affected by PD neurodegenerative changes. Defects in mitochondrial activity in the substantia nigra have been observed in the brain of patients affected by PD and substantia nigra lesions can induce the onset of a secondary parkinsonism. Thus, energy starvation and consequently metabolic impairment to dopaminergic neurons may be related to the onset of PD. On this line, we evaluated the effect of nutrient starvation, reproduced 'in vitro' by glucose deprivation (GD), in primary mesecephalic neuronal cultures and dopaminergic-differentiated SH-SY5Y cells, to evaluate if decreased glucose support to dopaminergic cells can lead to mitochondrial damage, neurodegeneration and alpha-syn misfolding. Furthermore, we investigated the effect of dopamine (DA) treatment in the presence of a DA-uptake inhibitor or of the D(2)/D(3) receptor (D(2)R/D(3)R) agonist quinpirole on GD-treated cells, to evaluate the efficacy of these therapeutic compounds. We found that GD induced the formation of fibrillary aggregated alpha-syn inclusions containing the DA transporter in dopaminergic cells. These alterations were accompanied by dopaminergic cell death and were exacerbated by DA overload. Conversely, the block of DA uptake and D(2)R/D(3)R agonist treatment exerted neuroprotective effects. These data indicate that glucose starvation is likely involved in the induction of PD-related pathological changes in dopaminergic neurons. These changes may be counteracted by the block of DA uptake and by dopaminergic agonist treatment.


Assuntos
Dopamina/metabolismo , Glucose/deficiência , Degeneração Neural/etiologia , Receptores de Dopamina D2/fisiologia , alfa-Sinucleína/metabolismo , Análise de Variância , Animais , Benzotiazóis , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Dopamina/farmacologia , Dopaminérgicos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Embrião de Mamíferos , Formazans/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Mesencéfalo/citologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transporte Proteico/efeitos dos fármacos , Sais de Tetrazólio/metabolismo , Tiazóis/metabolismo
16.
Eur J Neurosci ; 28(7): 1231-40, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18973551

RESUMO

Clinical improvements in Parkinson's disease produced by dopamine D3 receptor-preferring agonists have been related to their neuroprotective actions and, more recently, to their neuroregenerative properties. However, it is unclear whether dopamine agonists produce their neurotrophic effects by acting directly on receptors expressed by the mesencephalic dopaminergic neurons or indirectly on receptors expressed by astrocytes, via release of neurotrophic factors. In this study, we investigated the effects of the dopamine D3 receptor-preferring agonists quinpirole and 7-hydroxy-N,N-di-propyl-2-aminotetralin (7-OH-DPAT), as well as of the indirect agonist amphetamine, on dopaminergic neurons identified by tyrosine hydroxylase immunoreactivity (TH-IR). Experiments were performed on neuronal-enriched primary cultures containing less than 0.5% of astrocytes prepared from the mouse embryo mesencephalon. After 3 days of incubation, both quinpirole (1-10 microm) and 7-OH-DPAT (5-500 nm) dose-dependently increased the maximal dendrite length (P < 0.001), number of primary dendrites (P < 0.01) and [3H]dopamine uptake (P < 0.01) of TH-IR-positive mesencephalic neurons. Similar effects were observed with 10 microm amphetamine. All neurotrophic effects were blocked by the unselective D2/D3 receptor antagonist sulpiride (5 microm) and by the selective D3 receptor antagonist SB-277011-A at a low dose (50 nm). Quinpirole and 7-OH-DPAT also increased the phosphorylation of extracellular signal-regulated kinase (ERK) within minutes, an effect blocked by pretreatment with SB-277011-A. Inhibition of the D2/D3 receptor signalling pathway to ERK was obtained with PD98059, GF109203 or LY294002, resulting in blockade of neurotrophic effects. These data suggest that dopamine agonists increase dendritic arborizations of mesencephalic dopaminergic neurons via a direct effect on D2/D3 receptors, preferentially involving D3 receptor-dependent neurotransmission.


Assuntos
Dendritos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Neurogênese/fisiologia , Receptores de Dopamina D3/metabolismo , Anfetamina/farmacologia , Animais , Forma Celular/efeitos dos fármacos , Forma Celular/fisiologia , Células Cultivadas , Dendritos/efeitos dos fármacos , Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Mesencéfalo/citologia , Camundongos , Fatores de Crescimento Neural/farmacologia , Neurogênese/efeitos dos fármacos , Quimpirol/farmacologia , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/antagonistas & inibidores , Substância Negra/citologia , Substância Negra/embriologia , Substância Negra/metabolismo , Tetra-Hidronaftalenos/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
17.
Neuroreport ; 29(16): 1425-1430, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30211770

RESUMO

The mechanisms underlying the prolonged antidepressant effects after a single infusion of ketamine are only partially understood. Ketamine's half-life of ∼2 h cannot explain antidepressant effects that last for 1 week, suggesting the triggering of long-lasting neuroplasticity. Recent human pharmacokinetics (PK) data indicate that a ketamine metabolite, (2R,6R)-hydroxynorketamine (HNK), persists in the high submicromolar range for additional 6-12 h. As in rodents HNK can induce dendrite outgrowth through AMPA receptor-mediated mechanisms, in this work, we aimed to show that HNK produces similar effects in human neurons at concentrations and exposure time compatible with human PK after ketamine infusion. Human dopaminergic neurons were differentiated in-vitro from inducible pluripotent stem cells obtained from healthy donors. Exposure to submicromolar HNK for 6 h produced dendrite outgrowth when measured 3 days after exposure. This neuroplasticity was similar to that obtained with exposure to low micromolar concentrations of ketamine for 1 or 6 h. HNK and ketamine effects were blocked by pretreatment with the AMPA receptor antagonists NBQX and GYKI 52466, and by the mammalian target of rapamycin pathway blocker rapamycin. It is reasonable to conclude that the mechanistic similarity between ketamine and HNK and their diachronic brain exposure owing to the different plasma PK observed after single therapeutic ketamine infusion should contribute to the final sustained antidepressant action.


Assuntos
Dendritos/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Ketamina/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Receptores de AMPA/metabolismo , Células Cultivadas , Dopamina/metabolismo , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Mensageiro , Fatores de Tempo
18.
Neuroreport ; 29(8): 615-620, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29570499

RESUMO

Ketamine is a noncompetitive glutamate N-methyl-D-aspartic acid receptor antagonist. When acutely administered to rodents, it produces a rapid antidepressant effect. There is evidence that N-methyl-D-aspartic acid receptor blockade enhances glutamatergic transmission preferentially engaging α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors leading to mTOR (mammalian target of rapamycin) pathways activation, thus resulting into downstream neuroadaptive changes in limbic structures. Recent in-vitro data on primary neuronal cultures showed that ketamine activates mTOR also in dopaminergic neurons, and this activation depends on the presence of functional dopamine D3 receptors. The aim of this work was to study the in-vivo relevance of viable D3 receptors in mediating the effects of acute ketamine administration on the mTOR downstream substrate p70 ribosomal S6 kinase (p70S6K), an obligatory substrate for mTOR. We compared the effects of single ketamine 5 mg/kg, 10 mg/kg, or vehicle injection in wild-type and D3 receptor knockout mice. Animals were killed after 60 min, and their brains were processed for p-p70S6K immunohistochemistry. Ketamine increased p70S6K phosphorylation in prefrontal cortex, nucleus accumbens core and shell, ventral tegmental area, substantia nigra, hippocampal CA1, CA2, and CA3, and basolateral amygdala of wild-type mice but not in mutant mice. Our study demonstrates that ketamine-induced p70S6K phosphorylation is dependent on viable D3R expressed in most of limbic structures.


Assuntos
Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/farmacologia , Sistema Límbico/efeitos dos fármacos , Receptores de Dopamina D3/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Sistema Límbico/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Dopamina D3/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo
19.
Front Pharmacol ; 9: 572, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29910731

RESUMO

Midbrain dopamine (DA) neurons are considered a critical substrate for the reinforcing and sensitizing effects of nicotine and tobacco dependence. While the role of the α4 and ß2 subunit containing nicotinic acetylcholine receptors (α4ß2∗nAChRs) in mediating nicotine effects on DA release and DA neuron activity has been widely explored, less information is available on their role in the morphological adaptation of the DA system to nicotine, eventually leading to dysfunctional behaviors observed in nicotine dependence. In particular, no information is available on the role of α6∗nAChRs in nicotine-induced structural plasticity in rodents and no direct evidence exists regarding the occurrence of structural plasticity in human DA neurons exposed to nicotine. To approach this problem, we used two parallel in vitro systems, mouse primary DA neuron cultures from E12.5 embryos and human DA neurons differentiated from induced pluripotent stem cells (iPSCs) of healthy donors, identified using TH+ immunoreactivity. In both systems, nicotine 1-10 µM produced a dose-dependent increase of maximal dendrite length, number of primary dendrites, and soma size when measured after 3 days in culture. These effects were blocked by pretreatments with the α6∗nAChR antagonists α-conotoxin MII and α-conotoxin PIA, as well as by the α4ß2nAChR antagonist dihydro-ß-erythroidine (DHßE) in both mouse and human DA neurons. Nicotine was also ineffective when the primary DA neurons were obtained from null mutant mice for either the α6 subunit or both the α4 and α6 subunits of nAChR. When pregnant mice were exposed to nicotine from gestational day 15, structural plasticity was also observed in the midbrain DA neurons of postnatal day 1 offspring only in wild-type mice and not in both null mutant mice. This study confirmed the critical role of α4α6∗nAChRs in mediating nicotine-induced structural plasticity in both mouse and human DA neurons, supporting the translational relevance of neurons differentiated from human iPSCs for pharmacological studies.

20.
Eur J Hum Genet ; 26(12): 1824-1831, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30089821

RESUMO

The role of nicotinic acetylcholine receptors (nAChR) in nicotine dependence (ND) is well established; CHRNA7, encoding the α7 subunit, has a still uncertain role in ND, although it is implicated in a wide range of neuropsychiatric conditions. CHRFAM7A, a hybrid gene containing a partial duplication of CHRNA7, is possibly involved in modulating α7 nAChR function. The aim of this study was to investigate the role of CHRNA7 and CHRFAM7A genetic variants in ND and to test the hypothesis that α7 nAChR variation may modulate the efficacy of varenicline treatment in smoking cessation. We assessed CHRNA7 and CHRFAM7A copy number, CHRFAM7A exon 6 ∆2 bp polymorphism, and sequence variants in the CHRNA7 proximal promoter in an Italian sample of 408 treatment-seeking smokers. We conducted case-control and quantitative association analyses using two smoking measures (cigarettes per day, CPD, and Fagerström Test for Nicotine Dependence, FTND). Next, driven by the hypothesis that varenicline may exert some of its therapeutic effects through activation of α7 nAChRs, we restricted the analysis to a subgroup of 142 smokers who received varenicline treatment. The CHRNA7 promoter variant rs28531779 showed association with both smoking quantitative measures (FNTD p = 0.026, ß = 0.89, 95% CI 0.11-1.67; CPD p = 0.006, ß = 4.82 95% CI 1.42-8.22). Moreover, in the varenicline-treated subgroup we observed association of CHRFAM7A copy number with 6 months smoking abstinence (p = 0.035, OR = 3.18, 95% CI = 1.09-9.30). Thus, our study points to a possible role of genetic variation in CHRNA7 and CHRFAM7A in tobacco addiction mechanisms and response to varenicline treatment.


Assuntos
Variações do Número de Cópias de DNA , Polimorfismo de Nucleotídeo Único , Agentes de Cessação do Hábito de Fumar/uso terapêutico , Tabagismo/genética , Vareniclina/uso terapêutico , Receptor Nicotínico de Acetilcolina alfa7/genética , Adolescente , Adulto , Idoso , Resistência a Medicamentos/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Tabagismo/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA