Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Aging ; 3(8): 931-937, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37400721

RESUMO

Cognitive dysfunction in aging is a major biomedical challenge. Whether treatment with klotho, a longevity factor, could enhance cognition in human-relevant models such as in nonhuman primates is unknown and represents a major knowledge gap in the path to therapeutics. We validated the rhesus form of the klotho protein in mice showing it increased synaptic plasticity and cognition. We then found that a single administration of low-dose, but not high-dose, klotho enhanced memory in aged nonhuman primates. Systemic low-dose klotho treatment may prove therapeutic in aging humans.


Assuntos
Glucuronidase , Longevidade , Camundongos , Humanos , Animais , Idoso , Glucuronidase/metabolismo , Envelhecimento , Cognição , Primatas/metabolismo
2.
Psychopharmacology (Berl) ; 240(5): 1033-1048, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36961560

RESUMO

RATIONALE: Dopamine (DA) signaling through the D1 receptor has been shown to be integral to multiple aspects of cognition, including the core process of working memory. The discovery of positive allosteric modulators (PAMs) of the D1 receptor has enabled treatment modalities that may have alternative benefits to orthosteric D1 agonists arising from a synergism of action with functional D1 receptor signaling. OBJECTIVES: To investigate this potential, we have studied the effects of the novel D1 PAM DPTQ on a spatial delayed response working memory task in the rhesus monkey. Initial studies indicated that DPTQ binds to primate D1R with high affinity and selectivity and elevates spontaneous eye blink rate in rhesus monkeys in a dose-dependent manner consistent with plasma ligand exposures and central D1activation. RESULTS: Based on those results, DPTQ was tested at 2.5 mg/kg IM in the working memory task. No acute effect was observed 1 h after dosing, but performance was impaired 48 h later. Remarkably, this deficit was immediately followed by a significant enhancement in cognition over the next 3 days. In a second experiment in which DPTQ was administered on days 1 and 5, the early impairment was smaller and did not reach statistical significance, but statistically significant enhancement of performance was observed over the following week. Lower doses of 0.1 and 1.0 mg/kg were also capable of producing this protracted enhancement without inducing any transient impairment. CONCLUSIONS: DPTQ exemplifies a class of D1PAMs that may be capable of providing long-term improvements in working memory.


Assuntos
Memória de Curto Prazo , Receptores de Dopamina D1 , Animais , Receptores de Dopamina D1/metabolismo , Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Memória Espacial
3.
Front Pharmacol ; 11: 1005, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32733245

RESUMO

Selective activation of dopamine D1 receptors remains a promising pro-cognitive therapeutic strategy awaiting robust clinical investigation. PF-6142 is a key example from a recently disclosed novel series of non-catechol agonists and partial agonists of the dopamine D1/5 receptors (D1R) that exhibit pharmacokinetic (PK) properties suitable for oral delivery. Given their reported potential for functionally biased signaling compared to known catechol-based selective agonists, and the promising rodent PK profile of PF-6142, we utilized relevant in vivo assays in male rodents and male and female non-human primates (NHP) to evaluate the pharmacology of this new series. Studies in rodents showed that PF-6142 increased locomotor activity and prefrontal cortex acetylcholine release, increased time spent in wakefulness, and desynchronized the EEG, like known D1R agonists. D1R selectivity of PF-6142 was supported by lack of effect in D1R knock-out mice and blocked response in the presence of the D1R antagonist SCH-23390. Further, PF-6142 improved performance in rodent models of NMDA receptor antagonist-induced cognitive dysfunction, such as MK-801-disrupted paired-pulse facilitation, and ketamine-disrupted working memory performance in the radial arm maze. Similarly, PF-6142 reversed ketamine-induced deficits in NHP performing the spatial delayed recognition task. Of importance, PF-6142 did not alter the efficacy of risperidone in assays predictive of antipsychotic-like effect in rodents including pre-pulse inhibition and conditioned avoidance responding. These data support the continued development of non-catechol based D1R agonists for the treatment of cognitive impairment associated with brain disorders including schizophrenia.

4.
J Neurosci ; 34(32): 10592-602, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25100593

RESUMO

The elevation of kynurenic acid (KYNA) observed in schizophrenic patients may contribute to core symptoms arising from glutamate hypofunction, including cognitive impairments. Although increased KYNA levels reduce excitatory neurotransmission, KYNA has been proposed to act as an endogenous antagonist at the glycine site of the glutamate NMDA receptor (NMDAR) and as a negative allosteric modulator at the α7 nicotinic acetylcholine receptor. Levels of KYNA are elevated in CSF and the postmortem brain of schizophrenia patients, and these elevated levels of KYNA could contribute to NMDAR hypofunction and the cognitive deficits and negative symptoms associated with this disease. However, the impact of endogenously produced KYNA on brain function and behavior is less well understood due to a paucity of pharmacological tools. To address this issue, we identified PF-04859989, a brain-penetrable inhibitor of kynurenine aminotransferase II (KAT II), the enzyme responsible for most brain KYNA synthesis. In rats, systemic administration of PF-04859989 dose-dependently reduced brain KYNA to as little as 28% of basal levels, and prevented amphetamine- and ketamine-induced disruption of auditory gating and improved performance in a sustained attention task. It also prevented ketamine-induced disruption of performance in a working memory task and a spatial memory task in rodents and nonhuman primates, respectively. Together, these findings support the hypotheses that endogenous KYNA impacts cognitive function and that inhibition of KAT II, and consequent lowering of endogenous brain KYNA levels, improves cognitive performance under conditions considered relevant for schizophrenia.


Assuntos
Encéfalo/metabolismo , Cognição/fisiologia , Ácido Cinurênico/metabolismo , Esquizofrenia/líquido cefalorraquidiano , Esquizofrenia/patologia , Animais , Atenção/efeitos dos fármacos , Atenção/fisiologia , Inibidores Enzimáticos/farmacologia , Potenciais Evocados Auditivos/efeitos dos fármacos , Potenciais Evocados Auditivos/fisiologia , Feminino , Hipocampo/citologia , Humanos , Macaca mulatta , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Pirazóis/farmacologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Vigília
5.
J Nucl Med ; 54(11): 1962-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24115528

RESUMO

UNLABELLED: Neuroreceptor imaging in the nonhuman primate (NHP) is valuable for translational research approaches in humans. However, most NHP studies are conducted under anesthesia, which affects the interpretability of receptor binding measures. The aims of this study were to develop awake NHP imaging with minimal head restraint and to compare in vivo binding of the γ-aminobutyric acid type A (GABAA)-benzodiazepine radiotracer (11)C-flumazenil under anesthetized and awake conditions. We hypothesized that (11)C-flumazenil binding potential (BPND) would be higher in isoflurane-anesthetized monkeys. METHODS: The small animal PET scanner was fitted to a mechanical device that raised and tilted the scanner 45° while the awake NHP was tilted back 35° in a custom chair for optimal brain positioning, which required acclimation of the animals to the chair, touch-screen tasks, intravenous catheter insertion, and tilting. For PET studies, the bolus-plus-constant infusion method was used for (11)C-flumazenil administration. Two rhesus monkeys were scanned under the awake (n = 6 scans) and isoflurane-anesthetized (n = 4 scans) conditions. An infrared camera was used to track head motion during PET scans. Under the awake condition, emission and head motion-tracking data were acquired for 40-75 min after injection. Anesthetized monkeys were scanned for 90 min. Cortisol measurements were acquired during awake and anesthetized scans. Equilibrium analysis was used for both the anesthetized (n = 4) and the awake (n = 5) datasets to compute mean BPND images in NHP template space, using the pons as a reference region. The percentage change per minute in radioactivity concentration was calculated in high- and low-binding regions to assess the quality of equilibrium. RESULTS: The monkeys acclimated to procedures in the NHP chair necessary to perform awake PET imaging. Image quality was comparable between awake and anesthetized conditions. The relationship between awake and anesthetized values was BPND (awake) = 0.94 BPND (anesthetized) + 0.36 (r(2) = 0.95). Cortisol levels were significantly higher under the awake condition (P < 0.05). CONCLUSION: We successfully performed awake NHP imaging with minimal head restraint. There was close agreement in (11)C-flumazenil BPND values between awake and anesthetized conditions.


Assuntos
Benzodiazepinas/metabolismo , Encéfalo/diagnóstico por imagem , Flumazenil/metabolismo , Cabeça , Tomografia por Emissão de Pósitrons/métodos , Vigília , Ácido gama-Aminobutírico/metabolismo , Anestesia , Animais , Encéfalo/metabolismo , Radioisótopos de Carbono , Feminino , Macaca mulatta , Masculino , Tomografia por Emissão de Pósitrons/instrumentação , Restrição Física
7.
Biol Psychiatry ; 69(1): 12-8, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20965497

RESUMO

BACKGROUND: Nicotine improves cognition in humans and animal models of neuropsychiatric disorders. Here, we sought to establish whether selective stimulation of the neuronal nicotinic α7 receptor could improve spatial working memory in nonhuman primates. METHODS: Beginning with an estimated dose range from rodent studies, the dose of the α7 agonist AZD0328 was titrated for a significant impact on working memory in rhesus macaques after acute administration. After training to stability on the spatial delayed response task, subjects were administered AZD0328 (1.6 ng/kg-.48 mg/kg; intramuscular) or vehicle 30 min before cognitive testing. AZD0328 (1 ng/kg-1.0 µg/kg; intramuscular) was then administered in a repeated, intermittent ascending dose regimen where each dose was given in two bouts for 4 days with a 1-week washout in between bouts, followed by 2-week washout. RESULTS: Acute AZD0328 improved cognitive performance when the dose was titrated down to .0016 and .00048 mg/kg from a cognitively impairing dose of .48 mg/kg. In a subgroup, sustained enhancement of working memory was evident for 1 month or more after acute treatment. Immediate and sustained cognitive enhancement was also found during and after repeated administration of AZD0328 at .001 mg/kg. CONCLUSIONS: These findings demonstrate that extremely low doses of a nicotinic α7 agonist can have profound acute and long-lasting beneficial consequences for cognition, dependent upon the integrity of dorsolateral prefrontal cortex. Thus, the α7 receptor might have a fundamental role in the neural circuitry of working memory and in the synaptic plasticity upon which it might depend.


Assuntos
Furanos/farmacologia , Memória de Curto Prazo/efeitos dos fármacos , Nootrópicos/farmacologia , Desempenho Psicomotor/efeitos dos fármacos , Quinuclidinas/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Feminino , Furanos/administração & dosagem , Furanos/farmacocinética , Injeções Intramusculares , Macaca mulatta , Masculino , Nootrópicos/administração & dosagem , Nootrópicos/farmacocinética , Quinuclidinas/administração & dosagem , Quinuclidinas/farmacocinética , Receptor Nicotínico de Acetilcolina alfa7
8.
Pharmacol Biochem Behav ; 96(3): 325-32, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20600252

RESUMO

Neuroplasticity is a key factor in restoration of brain function following neuropathology associated with disease or drug exposure. Here we examined the potential for chronic treatment with the selective D1 receptor antagonist SCH39166 to reverse the profound and enduring cognitive impairment associated with amphetamine (AMPH) sensitization in the nonhuman primate and to stimulate re-growth of atrophied pyramidal dendrites in the dorsolateral prefrontal cortex of these animals. Four rhesus monkeys with sustained cognitive impairment (>1year following AMPH sensitization) were treated for up to 8months with SCH39166. Cognitive testing was performed before, during, and for up to 1(1/2) year following treatment. Significant improvement in working memory performance was observed only after cessation of the D1 antagonist treatment but then was sustained for the duration of the post-treatment testing period. Postmortem quantitative assessment of Golgi-impregnated pyramidal neurons in BA9 showed that apical dendritic length and trunk spine density were increased in D1 antagonist treated monkeys relative to AMPH-sensitized and AMPH-naïve monkeys. These findings, which suggest that the deleterious consequences of AMPH sensitization can be reversed by modulation of D1 receptor signaling, have implications for treating the underlying neural basis of cognitive deficits in both schizophrenia and substance abuse.


Assuntos
Anfetamina/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Cognição/efeitos dos fármacos , Antagonistas de Dopamina/farmacologia , Neurônios/efeitos dos fármacos , Receptores de Dopamina D1/antagonistas & inibidores , Animais , Benzazepinas/farmacologia , Dendritos/efeitos dos fármacos , Dendritos/ultraestrutura , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/ultraestrutura , Feminino , Macaca mulatta , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos
9.
Psychopharmacology (Berl) ; 210(3): 407-18, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20401749

RESUMO

RATIONALE: Ketamine has been used in humans to model cardinal symptoms of schizophrenia, including working memory impairments and behavioral disorganization. Translational studies with ketamine in nonhuman primates promise to extend the neurobiological understanding of this model. OBJECTIVES: By establishing the dose-dependent effects of ketamine on spatial working memory and behavior, we sought to test and compare the capacity of antipsychotic and procognitive agents to reverse these symptoms. METHODS: Behavioral observations were taken following administration of placebo/ketamine (0.1-1.7 mg/kg, intramuscularly) and animals were tested on the spatial delayed response task 15 min post-injection. Pretreatments with risperidone as well as full and partial D1 receptor agonists were tested for their ability to reverse ketamine-induced impairments. RESULTS: Ketamine (median 1.0 mg/kg) produced a profound cognitive impairment and behavioral sequelae reminiscent of positive and negative symptoms. Risperidone within the therapeutic dose range failed to antagonize behavioral or cognitive consequences of acute ketamine but A77636 (0.1 and 1 microg/kg) and SKF38393 (0.1 microg/kg-100 microg/kg) ameliorated the spatial working memory deficit. This effect of A77636 was blocked by the D1 receptor antagonist, SCH39166 (1 and 10 microg/kg). CONCLUSIONS: These findings establish a valuable ketamine platform relevant to the treatment of cognitive dysfunction in schizophrenia. The reversal of ketamine-induced working memory deficits by a D1 receptor agonist, but not a commonly prescribed atypical antipsychotic, provides behavioral evidence for significant D1/N-methyl-D: -aspartate receptor interactions in prefrontal dysfunction and concurs with suggestions that D1 agonists may be useful in the treatment of cognitive impairments in schizophrenia.


Assuntos
Agonistas de Dopamina/uso terapêutico , Ketamina/toxicidade , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Receptores de Dopamina D1/agonistas , Animais , Transtornos Cognitivos/tratamento farmacológico , Feminino , Humanos , Macaca mulatta , Masculino , Receptores de Dopamina D1/fisiologia , Esquizofrenia/tratamento farmacológico
10.
Biol Psychiatry ; 67(10): 998-1001, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20189164

RESUMO

BACKGROUND: Ketamine has been used to model cognitive and behavioral symptoms of schizophrenia. Current hypotheses state that inadequate glutamatergic transmission in schizophrenia leads to a deficiency in gamma-aminobutyric acid (GABA)ergic inhibitory mechanisms and treatment with a GABA type A receptor subunits alpha2/alpha3 (GABA(Aalpha2/3)) modulator improved working memory performance in a preliminary study in patients. Here, we used ketamine to impair spatial working memory and disrupt behavior to examine the capacity for the GABA(Aalpha2/3) agonist 7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine (TPA023) to reverse these symptoms. METHODS: Rhesus monkeys received TPA023 (.7, 2.0, and 5 mg/kg; by mouth) or vehicle 45 minutes before ketamine (1.0-1.7 mg/kg; intramuscular) or saline in a semirandomized Latin square design. Behavioral observations were acquired at approximately 5 minutes, and spatial delayed response performance was tested at 15 minutes postinjection. RESULTS: Ketamine produced a profound impairment in spatial working memory in association with the emergence of hallucinatory-like behaviors. TPA023 at all doses blocked ketamine's cognitive-impairing ability but did not influence the behavioral symptoms. CONCLUSIONS: Acute GABA(Aalpha2/3) agonist administration reverses the working memory deficits induced by ketamine in primates. This finding indicates that the consequences of N-methyl-D-aspartate deficiency on the function of prefrontal circuits involved in working memory can be completely overcome by acute enhancement of GABA signaling.


Assuntos
Agonistas de Receptores de GABA-A , Ketamina/antagonistas & inibidores , Transtornos da Memória/induzido quimicamente , Memória de Curto Prazo/efeitos dos fármacos , Piridazinas/farmacologia , Triazóis/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ketamina/farmacologia , Macaca mulatta
11.
Behav Brain Res ; 212(1): 41-8, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20347881

RESUMO

Working memory impairments are a core aspect of schizophrenia, yet current medicines do not address such cognitive dysfunction. We have developed a model of these working memory deficits by acutely disrupting glutamatergic synaptic transmission by administration of the N-methyl-d-aspartate (NMDA) antagonist ketamine in the nonhuman primate. The current studies evaluated the effect of positive allosteric modulators ("potentiators") of the alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors on the working memory and behavioral effects of ketamine. AMPA receptors mediate fast excitatory synaptic transmission throughout the brain and play a critical role in the activity-dependent regulation of NMDA receptors. We find that positive modulation of AMPA receptors with LY451646 (0.1-1.0mg/kg, SC) and structurally distinct PF-4778574 (0.01mg/kg, SC) robustly ameliorates ketamine-induced working memory impairments without altering behavioral effects of acute ketamine we consider related to positive- and negative-like symptoms. These results support AMPA receptor potentiators as a potential adjunctive treatment for cognitive impairment associated with schizophrenia (CIAS).


Assuntos
Agonistas de Aminoácidos Excitatórios/uso terapêutico , Ketamina , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/prevenção & controle , Memória de Curto Prazo/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Área Sob a Curva , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Agonistas de Aminoácidos Excitatórios/química , Macaca fascicularis , Macaca mulatta , Transtornos da Memória/sangue , Atividade Motora/efeitos dos fármacos , Testes Neuropsicológicos , Tempo de Reação/efeitos dos fármacos , Sulfonamidas/sangue , Sulfonamidas/química , Sulfonamidas/uso terapêutico , Tiofenos/farmacologia , Tiofenos/uso terapêutico , Fatores de Tempo
12.
Neuroreport ; 21(5): 390-4, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20186106

RESUMO

Glycine transporter inhibitors have recently been reported to improve symptoms in patients with schizophrenia. Here we used acute ketamine in the nonhuman primate to test the effectiveness of the novel glycine transporter inhibitor, PF-3463275, in a model of cognitive dysfunction relevant to schizophrenia. PF-3463275 (0.01-0.17 mg/kg; subcutaneously) or a vehicle was given before the administration of ketamine (median dose of 1.0 mg/kg intramuscularly) or placebo (saline). Ketamine induced hallucinatory-like behaviors that were not reversed by PF-3463275. In contrast, all doses of PF-3463275 alleviated the deficit in spatial working memory induced by ketamine. Theses findings build upon those in patients by providing translational support for targeting glycine transporter in adjunctive treatment for cognitive dysfunction in schizophrenia.


Assuntos
Compostos Azabicíclicos/uso terapêutico , Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Imidazóis/uso terapêutico , Transtornos da Memória/tratamento farmacológico , Memória de Curto Prazo/efeitos dos fármacos , Nootrópicos/uso terapêutico , Análise de Variância , Animais , Modelos Animais de Doenças , Feminino , Alucinações/induzido quimicamente , Alucinações/tratamento farmacológico , Macaca mulatta , Transtornos da Memória/induzido quimicamente , Testes Neuropsicológicos , Óxidos , Potássio , Esquizofrenia , Tantálio , Resultado do Tratamento
13.
Drug Discov Today ; 14(13-14): 690-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19460458

RESUMO

Current treatments for psychiatric disorders were developed with the aim of providing symptomatic relief rather than reversing underlying abnormalities in neuroplasticity or neurodevelopment that might contribute to psychiatric disorders. This review considers the possibility that psychiatric treatments might be developed that target neuroplasticity deficits or that manipulate neuroplasticity in novel ways. These treatments might not provide direct symptomatic relief. However, they might complement or enhance current pharmacotherapies and psychotherapies aimed at the prevention and treatment of psychiatric disorders. In considering neuroplasticity as a target for the treatment of psychiatric disorders, we build on exciting new findings in the areas of anxiety disorders, mood disorders, and schizophrenia.


Assuntos
Transtornos de Ansiedade/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Transtornos do Humor/metabolismo , Plasticidade Neuronal/fisiologia , Esquizofrenia/metabolismo , Animais , Transtornos de Ansiedade/tratamento farmacológico , Humanos , Transtornos do Humor/tratamento farmacológico , Estudos Multicêntricos como Assunto/métodos , Plasticidade Neuronal/efeitos dos fármacos , Preparações Farmacêuticas/administração & dosagem , Esquizofrenia/tratamento farmacológico
15.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(8): 1572-92, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17904719

RESUMO

Repeated, intermittent administration of psychomotor stimulants, or D1 agonists in dopamine-deficient states, induces behavioral sensitization, characterized by an enhanced response to a subsequent acute low dose challenge, which may be manifested in form of altered behavior or cognitive function. Amphetamine sensitization in the nonhuman primate encompasses profound and enduring changes to similar neuronal and neurochemical substrates that occur in rodents. The process of sensitization in the monkey also results in a long-lasting depression in baseline behavioral responding, as well as emergence of hallucinatory-like behaviors reminiscent of human psychosis in response to an acute challenge. Nonhuman primates show a reduction in spine density and dendritic length in prefrontal neurons and a marked reduction in basal dopamine turnover in both prefrontal cortex and striatum. A major hallmark of amphetamine sensitization in both nonhuman primates and rodents is the manifestation of deficits in executive function and working memory which rely upon the integrity of prefrontal cortex and thereby, may yield significant insights into the cognitive dysfunction associated with addiction. Together with evidence from human and rodent studies, it can be concluded that repeated exposure to psychomotor stimulants can lead to a corruption of neuroadaptive systems in the brain by an extraordinary influence on synaptic plasticity, learning, and memory. Actively harnessing this same process by repeated, intermittent D1 agonist administration may be the key to improved working memory and decision making in addiction and other dopamine dysfunctional states, such as schizophrenia.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Agonistas de Dopamina/farmacologia , Dopamina/fisiologia , Primatas/fisiologia , Animais , Estimulantes do Sistema Nervoso Central/toxicidade , Cognição/efeitos dos fármacos , Agonistas de Dopamina/toxicidade , Humanos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Receptores de Dopamina D1/efeitos dos fármacos
16.
Nat Neurosci ; 10(3): 376-84, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17277774

RESUMO

Dopamine (DA) D1 receptor (D1R) stimulation in prefrontal cortex (PFC) produces an 'inverted-U' dose-response, whereby either too little or too much D1R stimulation impairs spatial working memory. This response has been observed across species, including genetic linkages with human cognitive abilities, PFC activation states and DA synthesis. The cellular basis for the inverted U has long been sought, with in vitro intracellular recordings supporting a variety of potential mechanisms. The current study demonstrates that the D1R agonist inverted-U response can be observed in PFC neurons of behaving monkeys: low levels of D1R stimulation enhance spatial tuning by suppressing responses to nonpreferred directions, whereas high levels reduce delay-related firing for all directions, eroding tuning. These sculpting actions of D1R stimulation are mediated in monkeys and rats by cyclic AMP intracellular signaling. The evidence for an inverted U at the cellular level in behaving animals promises to bridge in vitro molecular analyses with human cognitive experience.


Assuntos
Memória de Curto Prazo/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/citologia , Receptores de Dopamina D1/fisiologia , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Comportamento Animal , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Dopaminérgicos/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Iontoforese/métodos , Macaca mulatta , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Testes Neuropsicológicos , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade , Tionucleotídeos/farmacologia
17.
Brain Cogn ; 63(2): 94-122, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17204357

RESUMO

The prefrontal cortex of the primate frontal lobes provides the capacity for judgment which can constantly adapt behavior in order to optimize its outcome. Adjudicating between long-term memory programs and prepotent responses, this capacity reviews all incoming information and provides an interpretation dependent on the events that have just occurred, the events that are predicted to happen, and the alternative response strategies that are available in the given situation. It has been theorized that this function requires two essential integrated components, a central executive which guides selective attention based on mechanisms of associative memory, as well as the second component, working memory buffers, in which information is held online, abstracted, and translated on a mental sketchpad of work in progress. In this review, we critically outline the evidence that the integration of these processes and, in particular, the induction and maintenance of persistent activity in prefrontal cortex and related networks, is dependent upon the interaction of dopamine D1 and glutamate NMDA receptor signaling at critical nodes within local circuits and distributed networks. We argue that this interaction is not only essential for representational memory, but also core to mechanisms of neuroadaptation and learning. Understanding its functional significance promises to reveal major new insights into prefrontal dysfunction in schizophrenia and, hence, to target a new generation of drugs designed to ameliorate the debilitating working memory deficits in this disorder.


Assuntos
Cognição/fisiologia , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Atenção/fisiologia , Humanos , Intenção , Memória de Curto Prazo/fisiologia
18.
Psychopharmacology (Berl) ; 174(1): 111-25, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15205882

RESUMO

BACKGROUND AND RATIONALE: Working memory performance is considered to be a core deficit in schizophrenia and the best predictor of social reintegration and propensity for relapse. This cardinal cognitive process is critical for human reasoning and judgment and depends upon the integrity of prefrontal function. Prefrontal dysfunction in schizophrenia has been linked to altered dopaminergic and glutamatergic transmission. However, to date, antipsychotics provide no substantial relief from the debilitating cognitive consequences of this disease. OBJECTIVES: This review examines the key rodent and non-human primate models for elucidating the neural mechanisms of working memory and their neuromodulation. We compare the physiology and pharmacology of working memory between the normal state and experimentally induced models of prefrontal dysfunction and evaluate their relevance for schizophrenia. RESULTS AND CONCLUSIONS: Rodent models have demonstrated the significance of aberrant dopaminergic and glutamatergic signaling in medial prefrontal cortex for working memory. However, there is some question as to the extent to which rodent tests of working memory tap into the same process that is compromised in schizophrenia. Non-human primates provide an unexcelled model for the study of influences on prefrontal function and working memory due to the high degree of homology between human and non-human primates in the relationship between prefrontal cortex and higher cognitive capacities. Moreover, non-human primate models of prefrontal dysfunction including amphetamine sensitization, subchronic phencyclidine, and neurodevelopmental insult are ideal for the analysis of novel compounds for the treatment of cognitive dysfunction in schizophrenia, thereby facilitating the translation between preclinical drug development and clinical trials.


Assuntos
Transtornos Cognitivos/etiologia , Memória de Curto Prazo/fisiologia , Modelos Animais , Esquizofrenia/fisiopatologia , Anfetamina , Animais , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Fenciclidina , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/induzido quimicamente
19.
Psychopharmacology (Berl) ; 174(1): 3-16, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15118803

RESUMO

BACKGROUND AND RATIONALE: Reinstatement of the function of working memory, the cardinal cognitive process essential for human reasoning and judgment, is potentially the most intractable problem for the treatment of schizophrenia. Since deficits in working memory are associated with dopamine dysregulation and altered D(1) receptor signaling within prefrontal cortex, we present the case for targeting novel drug therapies towards enhancing prefrontal D(1) stimulation for the amelioration of the debilitating cognitive deficits in schizophrenia. OBJECTIVES: This review examines the role of dopamine in regulating cellular and circuit function within prefrontal cortex in order to understand the significance of the dopamine dysregulation found in schizophrenia and related non-human primate models. By revealing the associations among prefrontal neuronal function, dopamine and D(1) signaling, and cognition, we seek to pinpoint the mechanisms by which dopamine modulates working memory processes and how these mechanisms may be exploited to improve cognitive function. RESULTS AND CONCLUSIONS: Dopamine deficiency within dorsolateral prefrontal cortex leads to abnormal recruitment of this region by cognitive tasks. Both preclinical and clinical studies have demonstrated a direct relationship between prefrontal dopamine function and the integrity of working memory, suggesting that insufficient D(1) receptor signaling in this region results in cognitive deficits. Moreover, working memory deficits can be ameliorated by treatments that augment D(1) receptor stimulation, indicating that this target presents a unique opportunity for the restoration of cognitive function in schizophrenia.


Assuntos
Transtornos Cognitivos/fisiopatologia , Receptores de Dopamina D1/fisiologia , Esquizofrenia/fisiopatologia , Animais , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Dopamina/metabolismo , Dopaminérgicos/farmacologia , Humanos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/complicações , Esquizofrenia/metabolismo , Transdução de Sinais/fisiologia
20.
J Neurosci ; 22(7): 2843-54, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11923449

RESUMO

Dorsolateral prefrontal cortex has an essential role in the cognitive process of working memory, dysfunction of which is considered to be a core deficit in schizophrenia. Although this cortical region is densely innervated with 5-HT2A receptors to which atypical antipsychotic drugs bind with high affinity, little is known of the influence of this serotonin receptor subtype on prefrontal function. We addressed this issue by examining the effects of iontophoresis of selective receptor ligands on prefrontal neurons possessing spatially tuned delay activity, or "memory fields," in monkeys performing a delayed-response task. Memory fields of putative pyramidal cells were attenuated by iontophoresis of 5-HT2A antagonists, which primarily produced a reduction in delay activity for preferred target locations. Conversely, 5-HT2A stimulation by alpha-methyl-5-HT or 5-HT itself, accentuated the spatial tuning of these neurons by producing a modest increase in activity for preferred target locations and/or a reduction in activity for nonpreferred locations. The agonist effects could be reversed by the selective antagonist MDL100,907, and were dose-dependent, such that high levels attenuated spatial tuning by profoundly reducing delay activity. A role for feedforward inhibitory circuitry in these effects was supported by the finding that 5-HT2A blockade also attenuated the memory fields of putative interneurons. We conclude that prefrontal 5-HT2A receptors have a hitherto unrecognized role in the cognitive function of working memory, which involves actions at both excitatory and inhibitory elements within local circuitry.


Assuntos
Memória de Curto Prazo/fisiologia , Neurônios/metabolismo , Córtex Pré-Frontal/fisiologia , Receptores de Serotonina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Cognição/efeitos dos fármacos , Cognição/fisiologia , Fixação Ocular/fisiologia , Iontoforese , Ligantes , Macaca mulatta , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estimulação Luminosa , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptor 5-HT2A de Serotonina , Movimentos Sacádicos/fisiologia , Antagonistas da Serotonina/administração & dosagem , Agonistas do Receptor de Serotonina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA