Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Aging ; 3(8): 931-937, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37400721

RESUMEN

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.


Asunto(s)
Glucuronidasa , Longevidad , Ratones , Humanos , Animales , Anciano , Glucuronidasa/metabolismo , Envejecimiento , Cognición , Primates/metabolismo
2.
Psychopharmacology (Berl) ; 240(5): 1033-1048, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36961560

RESUMEN

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.


Asunto(s)
Memoria a Corto Plazo , Receptores de Dopamina D1 , Animales , Receptores de Dopamina D1/metabolismo , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Memoria Espacial
3.
Front Pharmacol ; 11: 1005, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733245

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-25100593

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Cognición/fisiología , Ácido Quinurénico/metabolismo , Esquizofrenia/líquido cefalorraquídeo , Esquizofrenia/patología , Animales , Atención/efectos de los fármacos , Atención/fisiología , Inhibidores Enzimáticos/farmacología , Potenciales Evocados Auditivos/efectos de los fármacos , Potenciales Evocados Auditivos/fisiología , Femenino , Hipocampo/citología , Humanos , Macaca mulatta , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Pirazoles/farmacología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Vigilia
5.
J Nucl Med ; 54(11): 1962-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24115528

RESUMEN

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.


Asunto(s)
Benzodiazepinas/metabolismo , Encéfalo/diagnóstico por imagen , Flumazenil/metabolismo , Cabeza , Tomografía de Emisión de Positrones/métodos , Vigilia , Ácido gamma-Aminobutírico/metabolismo , Anestesia , Animales , Encéfalo/metabolismo , Radioisótopos de Carbono , Femenino , Macaca mulatta , Masculino , Tomografía de Emisión de Positrones/instrumentación , Restricción Física
7.
Biol Psychiatry ; 69(1): 12-8, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20965497

RESUMEN

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.


Asunto(s)
Furanos/farmacología , Memoria a Corto Plazo/efectos de los fármacos , Nootrópicos/farmacología , Desempeño Psicomotor/efectos de los fármacos , Quinuclidinas/farmacología , Receptores Nicotínicos/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Femenino , Furanos/administración & dosificación , Furanos/farmacocinética , Inyecciones Intramusculares , Macaca mulatta , Masculino , Nootrópicos/administración & dosificación , Nootrópicos/farmacocinética , Quinuclidinas/administración & dosificación , Quinuclidinas/farmacocinética , Receptor Nicotínico de Acetilcolina alfa 7
8.
Pharmacol Biochem Behav ; 96(3): 325-32, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20600252

RESUMEN

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.


Asunto(s)
Anfetamina/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Cognición/efectos de los fármacos , Antagonistas de Dopamina/farmacología , Neuronas/efectos de los fármacos , Receptores de Dopamina D1/antagonistas & inhibidores , Animales , Benzazepinas/farmacología , Dendritas/efectos de los fármacos , Dendritas/ultraestructura , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Femenino , Macaca mulatta , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos
9.
Psychopharmacology (Berl) ; 210(3): 407-18, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20401749

RESUMEN

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.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Ketamina/toxicidad , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Receptores de Dopamina D1/agonistas , Animales , Trastornos del Conocimiento/tratamiento farmacológico , Femenino , Humanos , Macaca mulatta , Masculino , Receptores de Dopamina D1/fisiología , Esquizofrenia/tratamiento farmacológico
10.
Biol Psychiatry ; 67(10): 998-1001, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20189164

RESUMEN

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.


Asunto(s)
Agonistas de Receptores de GABA-A , Ketamina/antagonistas & inhibidores , Trastornos de la Memoria/inducido químicamente , Memoria a Corto Plazo/efectos de los fármacos , Piridazinas/farmacología , Triazoles/farmacología , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ketamina/farmacología , Macaca mulatta
11.
Behav Brain Res ; 212(1): 41-8, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20347881

RESUMEN

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).


Asunto(s)
Agonistas de Aminoácidos Excitadores/uso terapéutico , Ketamina , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/prevención & control , Memoria a Corto Plazo/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Área Bajo la Curva , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Agonistas de Aminoácidos Excitadores/química , Macaca fascicularis , Macaca mulatta , Trastornos de la Memoria/sangre , Actividad Motora/efectos de los fármacos , Pruebas Neuropsicológicas , Tiempo de Reacción/efectos de los fármacos , Sulfonamidas/sangre , Sulfonamidas/química , Sulfonamidas/uso terapéutico , Tiofenos/farmacología , Tiofenos/uso terapéutico , Factores de Tiempo
12.
Neuroreport ; 21(5): 390-4, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20186106

RESUMEN

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.


Asunto(s)
Compuestos de Azabiciclo/uso terapéutico , Proteínas de Transporte de Glicina en la Membrana Plasmática/antagonistas & inhibidores , Imidazoles/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , Memoria a Corto Plazo/efectos de los fármacos , Nootrópicos/uso terapéutico , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Femenino , Alucinaciones/inducido químicamente , Alucinaciones/tratamiento farmacológico , Macaca mulatta , Trastornos de la Memoria/inducido químicamente , Pruebas Neuropsicológicas , Óxidos , Potasio , Esquizofrenia , Tantalio , Resultado del Tratamiento
13.
Drug Discov Today ; 14(13-14): 690-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19460458

RESUMEN

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.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Trastornos del Humor/metabolismo , Plasticidad Neuronal/fisiología , Esquizofrenia/metabolismo , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Humanos , Trastornos del Humor/tratamiento farmacológico , Estudios Multicéntricos como Asunto/métodos , Plasticidad Neuronal/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Esquizofrenia/tratamiento farmacológico
15.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(8): 1572-92, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17904719

RESUMEN

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.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Agonistas de Dopamina/farmacología , Dopamina/fisiología , Primates/fisiología , Animales , Estimulantes del Sistema Nervioso Central/toxicidad , Cognición/efectos de los fármacos , Agonistas de Dopamina/toxicidad , Humanos , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Receptores de Dopamina D1/efectos de los fármacos
16.
Brain Cogn ; 63(2): 94-122, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17204357

RESUMEN

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.


Asunto(s)
Cognición/fisiología , Corteza Prefrontal/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Atención/fisiología , Humanos , Intención , Memoria a Corto Plazo/fisiología
17.
Neuropsychopharmacology ; 32(4): 919-31, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16936713

RESUMEN

Amphetamine (AMPH) sensitization in the nonhuman primate induces persistent aberrant behaviors reminiscent of the hallmark symptoms of schizophrenia, including hallucinatory-like behaviors, psychomotor depression, and profound cognitive impairment. The present study examined whether AMPH sensitization induces similarly long-lasting morphologic alterations in prefrontal cortical pyramidal neurons. Three to 3(1/2) years postsensitization, sensitized, and AMPH-naïve control monkeys were killed. Blocks of prefrontal cortex were Golgi-impregnated for elucidation of pyramidal dendritic morphology in layers II/superficial III (II/IIIs), deep III, and V/VI. In AMPH-sensitized animals as compared to AMPH-naïve controls, pyramidal dendrites in layer II/IIIs exhibited reduced overall dendritic branching and reduced peak spine density (22%) on the apical trunk. Across all layers, the distance from soma to peak spine density along the apical trunk was decreased (126.38+/-7.65 mum in AMPH-sensitized compared to 162.98+/-7.26 microm in AMPH-naïve controls), and basilar dendritic length was reduced (32%). These findings indicate that chronic dopamine dysregulation, consequent to AMPH sensitization, results in enduring, atrophic changes in prefrontal pyramidal dendrites that resemble the pathologic alterations described in patients with schizophrenia and may contribute to the persistence of schizophrenia-like behavioral changes and cognitive dysfunction associated with sensitization. These findings may also provide key insights into the etiologic origin of the pronounced behavioral disturbances and cognitive dysfunction associated with schizophrenia.


Asunto(s)
Anfetamina/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Dendritas/efectos de los fármacos , Neuronas/citología , Corteza Prefrontal/citología , Animales , Diseño Asistido por Computadora , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Macaca mulatta
18.
Biol Psychiatry ; 57(7): 743-51, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15820231

RESUMEN

BACKGROUND: Amphetamine (AMPH) sensitization in monkeys produces long-lasting behavioral changes that model positive (hallucinatory-like behaviors) and negative (psychomotor depression) symptoms of schizophrenia. The extent to which this model produces the core deficit in schizophrenia--working memory impairment--is unknown. METHODS: Two groups of rhesus monkeys were sensitized to AMPH over 6 weeks. In one group, acquisition of cognitive tasks (delayed response, visual discrimination, delayed nonmatch-to-sample) was examined beginning 6+ months postsensitization. The second group was pretrained to stability on delayed response before sensitization. Regional postmortem concentrations of dopamine and its metabolites were examined in tissue from age-matched AMPH-naive and AMPH-sensitized monkeys using high-performance liquid chromatography with electrochemical detection (HPLC-ECD). RESULTS: The AMPH-sensitized monkeys were profoundly impaired in their ability to acquire cognitive tasks compared with AMPH-naïve monkeys. Pretrained monkeys showed impaired delayed response performance for several months following sensitization. Analysis by HPLC revealed that AMPH sensitization significantly reduced dopamine turnover in prefrontal cortex and striatum. CONCLUSIONS: Impairments in the acquisition and performance of spatial delayed response in association with reduced dopamine turnover in prefrontal cortex following AMPH sensitization provide further support for the relevance of this model to both the etiology and the treatment of cognitive dysfunction in schizophrenia.


Asunto(s)
Anfetamina/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos del Conocimiento/inducido químicamente , Dopamina/metabolismo , Corteza Prefrontal/efectos de los fármacos , Ácido 3,4-Dihidroxifenilacético/metabolismo , Anfetamina/efectos adversos , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/efectos adversos , Cromatografía Líquida de Alta Presión/métodos , Trastornos del Conocimiento/fisiopatología , Cuerpo Estriado/metabolismo , Aprendizaje Discriminativo/efectos de los fármacos , Electroquímica/métodos , Femenino , Ácido Homovanílico/metabolismo , Macaca mulatta , Masculino , Pruebas Neuropsicológicas/estadística & datos numéricos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Tiempo de Reacción/efectos de los fármacos , Recompensa , Factores de Tiempo , Percepción Visual/efectos de los fármacos
19.
Psychopharmacology (Berl) ; 174(1): 111-25, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15205882

RESUMEN

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.


Asunto(s)
Trastornos del Conocimiento/etiología , Memoria a Corto Plazo/fisiología , Modelos Animales , Esquizofrenia/fisiopatología , Anfetamina , Animales , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Humanos , Fenciclidina , Corteza Prefrontal/fisiopatología , Esquizofrenia/inducido químicamente
20.
Psychopharmacology (Berl) ; 174(1): 3-16, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15118803

RESUMEN

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.


Asunto(s)
Trastornos del Conocimiento/fisiopatología , Receptores de Dopamina D1/fisiología , Esquizofrenia/fisiopatología , Animales , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Dopaminérgicos/farmacología , Humanos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Esquizofrenia/complicaciones , Esquizofrenia/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...