Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Handb Exp Pharmacol ; 254: 399-415, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30701317

RESUMO

Conventional antidepressants increase the efflux of biogenic amine neurotransmitters (the monoamine hypothesis of depression) in the central nervous system (CNS) and are the principle drugs used to treat major depressive disorder (MDD). However, the lack of efficacy in some patients, the slow onset of action, and the side effect profiles of existing antidepressants necessitate the exploration of additional treatment options. The discovery of the nociceptin/orphanin FQ peptide NOP receptor (N/OFQ-NOP receptor) system and its characterization in preclinical biological and pharmacological stress-related conditions supports the potential antidepressant and anti-stress properties of a NOP receptor antagonist for the treatment of neurobehavioral disorders. BTRX-246040 (formerly LY2940094) was designed to test this hypothesis in the clinic. A small clinical proof of concept study demonstrated efficacy of BTRX-246040 in MDD patients. In this study, BTRX-246040 (40 mg, p.o.) significantly reduced negative bias as assessed by the facial recognition test within 1 week of treatment and decreased depression symptoms after 8 weeks. BTRX-246040 also reduced depression symptoms in a second trial with heavy alcohol drinkers. Given the comorbidity of MDD and alcohol use disorder, a compound with such effects in patients could be a valuable addition to the medications available. A proof of concept study showed efficacy of BTRX-246040 in reducing heavy drinking and increasing the probability of abstinence in individuals diagnosed with alcohol dependence. In addition, plasma levels of gamma-glutamyl transferase were decreased by BTRX-246040 compared to placebo control implying improvement in liver function. Collectively, the clinical data reviewed within this chapter suggest that BTRX-264040 functions to normalize dysfunction in reward circuits. The overall efficacy and safety of this compound with a novel mechanism of action are encouraging of further clinical development. BTRX-246040 is currently under development for MDD by BlackThorn Therapeutics.


Assuntos
Alcoolismo , Transtorno Depressivo Maior , Antagonistas de Entorpecentes/uso terapêutico , Peptídeos Opioides/farmacologia , Piranos/uso terapêutico , Compostos de Espiro/uso terapêutico , Alcoolismo/tratamento farmacológico , Humanos , Antagonistas de Entorpecentes/química , Peptídeos Opioides/química , Piranos/farmacologia , Compostos de Espiro/química , Compostos de Espiro/farmacologia
2.
Alzheimers Dement ; 14(2): 243-252, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28755839

RESUMO

INTRODUCTION: Our previous studies have shown that amyloid ß peptide (Aß) is subject to complement-mediated clearance from the peripheral circulation, and that this mechanism is deficient in Alzheimer's disease. The mechanism should be enhanced by Aß antibodies that form immune complexes (ICs) with Aß, and therefore may be relevant to current Aß immunotherapy approaches. METHODS: Multidisciplinary methods were employed to demonstrate enhanced complement-mediated capture of Aß antibody immune complexes compared with Aß alone in both erythrocytes and THP1-derived macrophages. RESULTS: Aß antibodies dramatically increased complement activation and opsonization of Aß, followed by commensurately enhanced Aß capture by human erythrocytes and macrophages. These in vitro findings were consistent with enhanced peripheral clearance of intravenously administered Aß antibody immune complexes in nonhuman primates. DISCUSSION: Together with our previous results, showing significant Alzheimer's disease deficits in peripheral Aß clearance, the present findings strongly suggest that peripheral mechanisms should not be ignored as contributors to the effects of Aß immunotherapy.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/imunologia , Anticorpos/sangue , Proteínas do Sistema Complemento/metabolismo , Eritrócitos/metabolismo , Imunoterapia/métodos , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/metabolismo , Animais , Adesão Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Fatores Imunológicos , Macaca fascicularis , Macrófagos/metabolismo , Masculino , Fagocitose , Células THP-1/metabolismo , Células THP-1/patologia
3.
Alzheimers Dement ; 13(12): 1397-1409, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28475854

RESUMO

INTRODUCTION: Although amyloid ß peptide (Aß) is cleared from the brain to cerebrospinal fluid and the peripheral circulation, mechanisms for its removal from blood remain unresolved. Primates have uniquely evolved a highly effective peripheral clearance mechanism for pathogens, immune adherence, in which erythrocyte complement receptor 1 (CR1) plays a major role. METHODS: Multidisciplinary methods were used to demonstrate immune adherence capture of Aß by erythrocytes and its deficiency in Alzheimer's disease (AD). RESULTS: Aß was shown to be subject to immune adherence at every step in the pathway. Aß dose-dependently activated serum complement. Complement-opsonized Aß was captured by erythrocytes via CR1. Erythrocytes, Aß, and hepatic Kupffer cells were colocalized in the human liver. Significant deficits in erythrocyte Aß levels were found in AD and mild cognitive impairment patients. DISCUSSION: CR1 polymorphisms elevate AD risk, and >80% of human CR1 is vested in erythrocytes to subserve immune adherence. The present results suggest that this pathway is pathophysiologically relevant in AD.


Assuntos
Doença de Alzheimer/sangue , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/sangue , Eritrócitos/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de Complemento/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/farmacologia , Animais , Estudos de Casos e Controles , Disfunção Cognitiva/patologia , Disfunção Cognitiva/fisiopatologia , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/ultraestrutura , Macaca fascicularis/sangue , Masculino , Testes de Estado Mental e Demência , Microscopia Eletrônica , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Receptores de Complemento/genética
4.
Handb Exp Pharmacol ; 228: 27-57, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25977079

RESUMO

Cognition is a complex brain function that represents processes such as learning and memory, attention, working memory, and executive functions amongst others. Impairments in cognition are prevalent in many neuropsychiatric and neurological disorders with few viable treatment options. The development of new therapies is challenging, and poor efficacy in clinical development continues to be one of the most consistent reasons compounds fail to advance, suggesting that traditional animal models are not predictive of human conditions and behavior. An effort to improve the construct validity of neuropsychological testing across species with the intent of facilitating therapeutic development has been strengthening over recent years. With an emphasis on understanding the underlying biology, optimizing the use of appropriate systems (e.g., transgenic animals) to model targeted disease states, and incorporating non-rodent species (e.g., non-human primates) that may enable a closer comparison to humans, an improvement in the translatability of the results will be possible. This chapter focuses on some promising translational cognitive paradigms for use in rodents, non-human primates, and humans.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Cognição/efeitos dos fármacos , Transtornos Mentais/tratamento farmacológico , Nootrópicos/uso terapêutico , Pesquisa Translacional Biomédica/métodos , Animais , Atenção/efeitos dos fármacos , Encéfalo/fisiopatologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/fisiopatologia , Transtornos Cognitivos/psicologia , Terapia Cognitivo-Comportamental/métodos , Modelos Animais de Doenças , Eletrocardiografia , Função Executiva/efeitos dos fármacos , Movimentos Oculares , Humanos , Imageamento por Ressonância Magnética , Memória/efeitos dos fármacos , Transtornos Mentais/diagnóstico , Transtornos Mentais/fisiopatologia , Transtornos Mentais/psicologia , Testes Neuropsicológicos , Valor Preditivo dos Testes
5.
Neuropharmacology ; 257: 110037, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38876309

RESUMO

Kappa opioid receptors (KORs) are implicated in the pathophysiology of various psychiatric and neurological disorders creating interest in targeting the KOR system for therapeutic purposes. Accordingly, navacaprant (NMRA-140) is a potent, selective KOR antagonist being evaluated as a treatment for major depressive disorder. In the present report, we have extended the pharmacological characterization of navacaprant by further demonstrating its selective KOR antagonist properties and confirming its lack of agonist activity at KORs and related targets involved in opioid-related abuse. Using CHO-K1 cells expressing human KOR, mu (MOR), or delta (DOR) opioid receptors, navacaprant demonstrated selective antagonist properties at KOR (IC50 = 0.029 µM) versus MOR (IC50 = 3.3 µM) and DOR (IC50 > 10 µM) in vitro. In vivo, navacaprant (10-30 mg/kg, i.p.) dose-dependently abolished KOR-agonist induced analgesia in the mouse tail-flick assay. Additionally, navacaprant (10, 30 mg/kg, p.o.) significantly reduced KOR agonist-stimulated prolactin release in mice and rats, confirming KOR antagonism in vivo. Navacaprant showed no agonist activity at any opioid receptor subtype (EC50 > 10 µM) in vitro and exhibited no analgesic effect in the tail-flick assays at doses ≤100 mg/kg, p.o. thereby confirming a lack of opioid receptor agonist activity in vivo. Importantly, navacaprant did not alter extracellular dopamine concentrations in the nucleus accumbens shell of freely-moving rats following doses ≤100 mg/kg, p.o., whereas morphine (10, 20 mg/kg, i.p.) significantly increased dopamine levels. These results demonstrate that navacaprant is a KOR-selective antagonist with no pharmacological properties implicated in opioid-related abuse.


Assuntos
Analgésicos Opioides , Cricetulus , Receptores Opioides kappa , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Animais , Células CHO , Humanos , Masculino , Camundongos , Ratos , Analgésicos Opioides/farmacologia , Cricetinae , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Antagonistas de Entorpecentes/farmacologia , Relação Dose-Resposta a Droga , Ratos Sprague-Dawley , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Camundongos Endogâmicos C57BL , Dopamina/metabolismo
6.
Neurobiol Stress ; 21: 100493, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36532373

RESUMO

Extensive preclinical and emerging clinical evidence point to an involvement of the kappa opioid receptor (KOR) in brain networks that promotes neurobehavioral stability. KOR expression in mesolimbic and mesocortical pathways has been the basis for characterizing the role of this receptor system in regulating motivation and emotion; however, the involvement of the KOR system in higher-order executive processes such as working memory (WM) is not well-understood. WM is readily impaired with uncontrollable stress exposure and is dysregulated in many neurobehavioral disorders. To empirically evaluate the role of the KOR system on WM performance, we administered a selective KOR antagonist, NMRA-140 (0, 0.1, 0.3, 1.0 mg/kg, intramuscular) to monkeys under both stress and non-stress conditions. In this study, NMRA-140 was co-administered with FG7142, a benzodiazepine inverse agonist, known to produce a mild stress response and to impair WM function in monkeys. NMRA-140 protected WM performance from the detrimental effects of FG7142-induced stress and exhibited no significant effect under non-stress conditions. Collectively, these data highlight the functional influence of the KOR system in mediating stress-induced dysfunction of executive processes and suggest that modulating KOR activity could offer therapeutic benefit in stress-related neurobehavioral disorders.

7.
J Pharmacol Exp Ther ; 336(1): 242-53, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20959364

RESUMO

Neuronal nicotinic α7 acetylcholine receptors (α7nAChRs) are expressed primarily in the brain and are implicated in modulating many cognitive functions (e.g., attention, working and episodic memory). Not surprisingly, much effort has been committed to the development of molecules acting at α7nAChRs as potential therapies for a variety of central nervous system diseases (e.g., Alzheimer's). N-[(3S)-1-azabicyclo[2.2.2]oct-3-yl]-1H-indazole-3-carboxamide hydrochloride (RG3487) binds potently to the human α7nAChR (K(i) = 6 nM), in which it acts as a partial agonist (63-69% of acetylcholine) as assessed by whole-cell patch-clamp recordings in both oocytes and QM7 cell lines. RG3487 activates human α7nAChRs with an EC(50) of 0.8 µM (oocytes) and 7.7 µM (QM7 cells). RG3487 also exhibits antagonist properties at the serotonin 3 receptor [IC(50) = 2.8 nM (oocytes), 32.7 nM (N1E-115 cells)]. In vivo, RG3487 improved object recognition memory in rats after acute [minimally effective dose (MED) 1.0 mg/kg p.o.] or repeated (10 day) administration at brain and plasma concentrations in the low-nanomolar range. Spatial learning deficits in age-impaired rats were reversed after RG3487 administration (MED: 0.03 mg/kg i.p.) as evaluated in the Morris water maze task. In the prepulse inhibition (PPI) of startle model of sensorimotor gating, RG3487 improved apomorphine-induced deficits in PPI performance (MED: 0.03 mg/kg i.p.) and reversed phencyclidine-induced impairments in an attentional set-shifting model of executive function (MED: ≤0.03 mg/kg i.p.). Cumulative evidence from these studies indicates RG3487 is a novel and potent α7nAChR partial agonist that improves cognitive performance and sensorimotor gating.


Assuntos
Compostos Bicíclicos com Pontes/farmacologia , Cognição/fisiologia , Agonismo Parcial de Drogas , Indazóis/farmacologia , Agonistas Nicotínicos/farmacologia , Receptores Nicotínicos/fisiologia , Filtro Sensorial/fisiologia , Animais , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Linhagem Celular Tumoral , Cognição/efeitos dos fármacos , Feminino , Humanos , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Long-Evans , Ratos Sprague-Dawley , Ratos Wistar , Filtro Sensorial/efeitos dos fármacos , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7
8.
Sci Rep ; 11(1): 7700, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33833333

RESUMO

GABAA-α5 subunit-containing receptors have been shown to play a key modulatory role in cognition and represent a promising drug target for cognitive dysfunction, as well as other disorders. Here we report on the preclinical and early clinical profile of a novel GABAA-α5 selective negative allosteric modulator (NAM), basmisanil, which progressed into Phase II trials for intellectual disability in Down syndrome and cognitive impairment associated with schizophrenia. Preclinical pharmacology studies showed that basmisanil is the most selective GABAA-α5 receptor NAM described so far. Basmisanil bound to recombinant human GABAA-α5 receptors with 5 nM affinity and more than 90-fold selectivity versus α1, α2, and α3 subunit-containing receptors. Moreover, basmisanil inhibited GABA-induced currents at GABAA-α5 yet had little or no effect at the other receptor subtypes. An in vivo occupancy study in rats showed dose-dependent target engagement and was utilized to establish the plasma exposure to receptor occupancy relationship. At estimated receptor occupancies between 30 and 65% basmisanil attenuated diazepam-induced spatial learning impairment in rats (Morris water maze), improved executive function in non-human primates (object retrieval), without showing anxiogenic or proconvulsant effects in rats. During the Phase I open-label studies, basmisanil showed good safety and tolerability in healthy volunteers at maximum GABAA-α5 receptor occupancy as confirmed by PET analysis with the tracer [11C]-Ro 15-4513. An exploratory EEG study provided evidence for functional activity of basmisanil in human brain. Therefore, these preclinical and early clinical studies show that basmisanil has an ideal profile to investigate potential clinical benefits of GABAA-α5 receptor negative modulation.


Assuntos
Agonistas de Receptores de GABA-A/farmacologia , Receptores de GABA-A/efeitos dos fármacos , Regulação Alostérica , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Células HEK293 , Voluntários Saudáveis , Humanos , Aprendizagem/efeitos dos fármacos , Macaca fascicularis , Tomografia por Emissão de Pósitrons , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Xenopus laevis
9.
Curr Top Behav Neurosci ; 45: 1-28, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32451956

RESUMO

Since its identification over a hundred years ago, the neurotransmitter acetylcholine (ACh) has proven to play an essential role in supporting many diverse functions. Some well-characterized functions include: chemical transmission at the neuromuscular junction; autonomic function in the peripheral nervous system; and, sustained attention, sleep/wake regulation, and learning and memory within the central nervous system. Within the brain, major cholinergic projection pathways from the basal forebrain and the brainstem support these centrally mediated processes, and dysregulation of the cholinergic system is implicated in cognitive decline associated with aging and dementias including Alzheimer's disease. ACh exerts its effects by binding to two different membrane-bound receptor classes: (1) G­protein coupled muscarinic acetylcholine receptors (mAChRs), and (2) ligand-gated nicotinic acetylcholine receptors (nAChRs). These receptor systems are described in detail within this chapter along with discussion on the successes and failures of synthetic ligands designed to selectively target receptor subtypes for treating brain disorders. New molecular approaches and advances in our understanding of the target biology combined with opportunities to re-purpose existing cholinergic drugs for new indications continue to highlight the exciting opportunities for modulating this system for therapeutic purposes.


Assuntos
Doença de Alzheimer , Receptores Nicotínicos , Acetilcolina , Encéfalo/metabolismo , Colinérgicos/uso terapêutico , Humanos , Receptores Nicotínicos/metabolismo
10.
PLoS One ; 15(12): e0232864, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33373369

RESUMO

Activation of the kappa opioid receptor (KOR) contributes to the aversive properties of stress, and modulates key neuronal circuits underlying many neurobehavioral disorders. KOR agonists directly inhibit ventral tegmental area (VTA) dopaminergic neurons, contributing to aversive responses (Margolis et al. 2003, 2006); therefore, selective KOR antagonists represent a novel therapeutic approach to restore circuit function. We used whole cell electrophysiology in acute rat midbrain slices to evaluate pharmacological properties of four novel KOR antagonists: BTRX-335140, BTRX-395750, PF-04455242, and JNJ-67953964. Each compound concentration-dependently reduced the outward current induced by the KOR selective agonist U-69,593. BTRX-335140 and BTRX-395750 fully blocked U-69,593 currents (IC50 = 1.2 ± 0.9 and 1.2 ± 1.3 nM, respectively). JNJ-67953964 showed an IC50 of 3.0 ± 4.6 nM. PF-04455242 exhibited partial antagonist activity asymptoting at 55% blockade (IC50 = 6.7 ± 15.1 nM). In 3/8 of neurons, 1 µM PF-04455242 generated an outward current independent of KOR activation. BTRX-335140 (10 nM) did not affect responses to saturating concentrations of the mu opioid receptor (MOR) agonist DAMGO or the delta opioid receptor (DOR) agonist DPDPE, while JNJ-67953964 (10 nM) partially blocked DAMGO and DPDPE responses. Importantly, BTRX-335140 (10 nM) rapidly washed out with complete recovery of U-69,593 responses within 10 min. Collectively, we show electrophysiological evidence of key differences amongst KOR antagonists that could impact their therapeutic potential and have not been observed using recombinant systems. The results of this study demonstrate the value of characterizing compounds in native neuronal tissue and within circuits implicated in the neurobehavioral disorders of interest.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Receptores Opioides kappa/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Benzamidas/farmacologia , Compostos de Bifenilo/farmacologia , Neurônios Dopaminérgicos/metabolismo , Eletrofisiologia , D-Penicilina (2,5)-Encefalina/farmacologia , Masculino , Mesencéfalo/metabolismo , Antagonistas de Entorpecentes/farmacologia , Oxidiazóis/farmacologia , Técnicas de Patch-Clamp/métodos , Piperidinas/farmacologia , Pirrolidinas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Sulfonamidas/farmacologia , Área Tegmentar Ventral/efeitos dos fármacos
11.
eNeuro ; 7(5)2020.
Artigo em Inglês | MEDLINE | ID: mdl-32747458

RESUMO

The neuropeptide nociceptin/orphanin FQ (N/OFQ) can be released by stressors and is associated with disorders of emotion regulation and reward processing. N/OFQ and its receptor, NOP, are enriched in dopaminergic pathways, and intra-ventricular agonist delivery decreases dopamine levels in the dorsal striatum, nucleus accumbens (NAc), and ventral tegmental area (VTA). We used whole-cell electrophysiology in acute rat midbrain slices to investigate synaptic actions of N/OFQ. N/OFQ was primarily inhibitory, causing outward currents in both immunocytochemically identified dopaminergic (tyrosine hydroxylase positive (TH(+))) and non-dopaminergic (TH(-)) VTA neurons; effect at 1 µm: 20 ± 4 pA. Surprisingly, this effect was mediated by augmentation of postsynaptic GABAAR currents, unlike the substantia nigra pars compacta (SNc), where the N/OFQ-induced outward currents were K+ channel dependent. A smaller population, 17% of all VTA neurons, responded to low concentrations of N/OFQ with inward currents (10 nm: -11 ± 2 pA). Following 100 nm N/OFQ, the response to a second N/OFQ application was markedly diminished in VTA neurons (14 ± 10% of first response) but not in SNc neurons (90 ± 20% of first response). N/OFQ generated outward currents in medial prefrontal cortex (mPFC)-projecting VTA neurons, but inward currents in a subset of posterior anterior cingulate cortex (pACC)-projecting VTA neurons. While N/OFQ inhibited NAc-projecting VTA cell bodies, it had little effect on electrically or optogenetically evoked terminal dopamine release in the NAc measured ex vivo with fast scan cyclic voltammetry (FSCV). These results extend our understanding of the N/OFQ system in brainstem circuits implicated in many neurobehavioral disorders.


Assuntos
Receptores Opioides , Área Tegmentar Ventral , Animais , Dopamina , Peptídeos Opioides , Ratos , Receptores Opioides/metabolismo , Área Tegmentar Ventral/metabolismo , Nociceptina
12.
Neuropsychopharmacology ; 44(8): 1485-1493, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30954024

RESUMO

Trace amine-associated receptor 1 (TAAR1) is a G-protein coupled receptor with affinity for the trace amines. TAAR1 agonists have pro-cognitive, antidepressant-, and antipsychotic-like properties in both rodents and non-human primates (NHPs). TAAR1 agonism also increases wakefulness and suppresses rapid-eye movement (REM) sleep in mice and rats and reduces cataplexy in two mouse models of narcolepsy. We investigated the effects of TAAR1 agonism in Cynomolgus macaques, a diurnal species that exhibits consolidated night-time sleep, and evaluated the effects of TAAR1 agonists on cognition using a working memory (WM) paradigm in this species. Adult male Cynomolgus macaques (n = 6) were surgically implanted to record the electroencephalogram (EEG), electromyogram, and locomotor activity (LMA) and the efficacy of the TAAR1 partial agonist RO5263397 (0.1,1,10 mg/kg, p.o.) on sleep/wake, EEG spectra, and LMA was determined. In a second experiment, the acute effects of RO5263397 (0.1,1,10 mg/kg, p.o.) were assessed on a delayed-match-to-sample test of WM in adult male macaques (n = 7). RO5263397 (10 mg/kg) administered at lights off, when sleep pressure was high, promoted wakefulness and reduced both REM and non-REM sleep without inducing hyperlocomotion. RO5263397 (10 mg/kg) also increased delta/theta activity during all vigilance states. RO5263397 had no effect on WM at either short (2 sec) or long (10 sec) delay intervals. The wake-enhancing and REM-suppressing effects of R05263397 shown here in a diurnal primate are consistent with previous results in nocturnal rodents. These effects and the associated alterations in EEG spectra occurred without inducing hyperlocomotion or affecting WM, encouraging further study of TAAR1 agonists as potential narcolepsy therapeutics.


Assuntos
Cognição/efeitos dos fármacos , Oxazóis/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Vigília/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Eletroencefalografia/efeitos dos fármacos , Macaca fascicularis , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos
13.
Artigo em Inglês | MEDLINE | ID: mdl-31236518

RESUMO

Most preclinical sleep studies are conducted in nocturnal rodents that have fragmented sleep in comparison to humans who are primarily diurnal, typically with a consolidated sleep period. Consequently, we sought to define basal sleep characteristics, sleep/wake architecture and electroencephalographic (EEG) activity in a diurnal non-human primate (NHP) to evaluate the utility of this species for pharmacological manipulation of the sleep/wake cycle. Adult, 9-11 y.o. male cynomolgus macaques (n = 6) were implanted with telemetry transmitters to record EEG and electromyogram (EMG) activity and Acticals to assess locomotor activity under baseline conditions and following injections either with vehicle or the caffeine (CAF; 10 mg/kg, i.m.) prior to the 12 h dark phase. EEG/EMG recordings (12-36 h in duration) were analyzed for sleep/wake states and EEG spectral composition. Macaques exhibited a sleep state distribution and architecture similar to previous NHP and human sleep studies. Acute administration of CAF prior to light offset enhanced wakefulness nearly 4-fold during the dark phase with consequent reductions in both NREM and REM sleep, decreased slow wave activity during wakefulness, and increased higher EEG frequency activity during NREM sleep. Despite the large increase in wakefulness and profound reduction in sleep during the dark phase, no sleep rebound was observed during the 24 h light and dark phases following caffeine administration. Cynomolgus macaques show sleep characteristics, EEG spectral structure, and respond to CAF in a similar manner to humans. Consequently, monitoring EEG/EMG by telemetry in this species may be useful both for basic sleep/wake studies and for pre-clinical assessments of drug-induced effects on sleep/wake.

14.
Eur J Neurosci ; 28(3): 625-32, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18702734

RESUMO

Elevation of intracellular cyclic adenosine monophosphate (cAMP) concentrations and subsequent regulation of downstream target gene expression through phosphorylation of cAMP-responsive element binding protein (CREB) is hypothesized to underlie the mechanism(s) of long-term memory (LTM) formation. The phosphodiesterase 4 (PDE4) enzyme family is believed to play a key role in LTM by regulating cAMP levels. Thus far, four PDE4 isoforms have been identified (PDE4A, B, C and D); however, the requisite involvement of each of these isoforms in mediating LTM has yet to be elucidated. In the present study, genetic knockout mice were used to investigate the involvement of the PDE4D isoform in both in vitro and in vivo models of learning and memory. Hippocampal synaptic transmission measured electrophysiologically in CA1 slice preparations was similar between wild-type and PDE4D (-/-) mice yet, relative to wild-type controls, knockout mice displayed enhanced early long-term potentiation (LTP) following multiple induction protocols. Interestingly, the PDE4D (-/-) animals exhibited significant behavioral deficits in associative learning using a conditioned fear paradigm as compared with control littermates. The impairment in fear conditioning observed in the PDE4D (-/-) mice could not be attributed to differences in acquisition of the task, alterations in locomotor activity or effects on shock sensitivity. Overall, the in vitro and in vivo alterations in synaptic plasticity observed in the PDE4D (-/-) mice may be explained by adaptive responses occurring throughout development, and suggest that the PDE4D isoform may be an important mediator of LTM formation.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Isoenzimas/metabolismo , Aprendizagem/fisiologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Animais , Comportamento Animal/fisiologia , Condicionamento Clássico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Eletrofisiologia , Medo/fisiologia , Isoenzimas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Plasticidade Neuronal/fisiologia , Transmissão Sináptica/fisiologia
16.
Neuropsychopharmacology ; 41(5): 1319-28, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26354045

RESUMO

A growing body of evidence indicates that neuronal oscillations in the gamma frequency range (30-80 Hz) are disturbed in schizophrenic patients during cognitive processes and may represent an endophenotype of the disease. N-methyl-D-aspartate (NMDA) receptor antagonists have been used experimentally to induce schizophrenia-like symptoms including cognitive deficits in animals and humans. Here we characterized neuronal oscillations and event-related potentials (ERPs) in Cynomolgus macaques fully trained to perform a continuous performance test (CPT) in the presence and absence of the NMDA antagonist phencyclidine (PCP). Macaques (n=8) were trained to touch 'target' stimuli and ignore 'distractor' stimuli presented randomly on a touchscreen. Subsequently, all subjects were implanted with epidural EEG electrodes over frontal (FC) and parietal cortices (PC) and later tested under vehicle (saline, i.m.) or acute PCP (0.1-0.3 mg/kg, i.m.) conditions. Compared with vehicle treatment, PCP produced a significant dose-dependent decrease in CPT performance accuracy and increased reaction times. Furthermore, PCP elevated the amplitudes of 'low' (30-50 Hz) and 'high' (51-80 Hz) gamma oscillations in FC and PC around target presentations for all correct responses. The CPT accuracy was inversely correlated with the gamma band amplitude in the presence of PCP. Additionally, PCP delayed the N100 peak latency in FC, and prolonged and suppressed the cognitively relevant P300 component of mean ERPs in FC and PC, respectively. The NMDA receptor antagonist-induced alteration in neuronal oscillations and ERPs may contribute to the observed cognitive deficits in macaques, and enhance our understanding of EEG recordings as a translatable biomarker.


Assuntos
Atenção/fisiologia , Lobo Frontal/fisiologia , Ritmo Gama , Lobo Parietal/fisiologia , Fenciclidina/administração & dosagem , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/fisiologia , Esquizofrenia/fisiopatologia , Animais , Atenção/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletroencefalografia , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Lobo Frontal/efeitos dos fármacos , Ritmo Gama/efeitos dos fármacos , Macaca fascicularis , Masculino , Lobo Parietal/efeitos dos fármacos
17.
J Neurosci ; 22(24): 10883-90, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12486182

RESUMO

The cAMP response element-binding protein (CREB) is a critical integrator of neural plasticity that is responsive in a brain region-specific manner to a variety of environmental and pharmacological stimuli, including widely prescribed antidepressant medications. We developed inducible transgenic lines of mice that express either CREB or a dominant-negative mutant of CREB (mCREB) in forebrain regions and used these mice to determine the functional significance of this transcription factor in the learned helplessness paradigm, a behavioral model of depression. We also use a complementary viral-mediated gene transfer approach to directly test the effect of mCREB in the nucleus accumbens, a brain region important for motivation and reward. The results demonstrate that blockade of CREB by overexpression of mCREB in transgenic mice or by viral expression of mCREB in the nucleus accumbens produces an antidepressant-like effect, whereas overexpression of CREB in transgenic mice results in the opposite phenotype. In addition, mCREB expression was colocalized with and decreased the expression of prodynorphin in nucleus accumbens medium spiny neurons, and antagonism of dynorphin in the nucleus accumbens was sufficient to produce an antidepressant-like effect similar to that observed after blockade of CREB. Together, the results demonstrate that nucleus accumbens CREB-dynorphin influence behavior in the learned helplessness model and suggest that this signaling cascade may contribute to symptoms of depression.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Depressão/etiologia , Dinorfinas/antagonistas & inibidores , Naltrexona/análogos & derivados , Núcleo Accumbens/fisiologia , Animais , Antidepressivos , Comportamento Animal , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Depressão/psicologia , Encefalinas/análise , Encefalinas/imunologia , Encefalinas/metabolismo , Desamparo Aprendido , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Naltrexona/farmacologia , Prosencéfalo/metabolismo , Precursores de Proteínas/análise , Precursores de Proteínas/imunologia , Precursores de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides kappa/antagonistas & inibidores
18.
Int Rev Neurobiol ; 124: 79-111, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26472526

RESUMO

Schizophrenia is a lifelong disease, the burden of which is often underestimated. Characterized by positive (e.g., hallucinations) and negative (e.g., avolition, amotivation) symptoms, schizophrenia is also accompanied with profound impairments in cognitive function that progress throughout the development of the disease. Although treatment with antipsychotic medications can effectively dampen some of the positive symptoms, these medications largely fail to reverse cognitive deficits or to mitigate negative symptoms. With a worldwide prevalence of approximately 1%, schizophrenia remains a large unmet medical need that stands to benefit greatly from (1) continued research to better understand the biological underpinnings of the disease and (2) the targeted development of novel therapeutics to improve the lives of those affected individuals. Improvements in our understanding of the neuronal networks associated with schizophrenia as well as progress in identifying genetic risk factors and environmental conditions that may predispose individuals to developing the disease are advancing new strategies to study and treat it. Herein, we review the evidence that supports the role of α7 nicotinic acetylcholine receptors in the central nervous system and why these receptors constitute a promising target to treat some of the prominent symptoms of schizophrenia.


Assuntos
Colinérgicos/uso terapêutico , Esquizofrenia/metabolismo , Esquizofrenia/terapia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Humanos , Pesquisa Translacional Biomédica
19.
Biol Psychiatry ; 56(3): 151-60, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15271583

RESUMO

BACKGROUND: Chronic antidepressant administration increases the cyclic adenosine monophosphate response element binding protein (CREB) in the amygdala, a critical neural substrate involved in the physiologic responses to stress, fear, and anxiety. METHODS: To determine the role of CREB in the amygdala in animal models of depression and anxiety, a viral gene transfer approach was used to selectively express CREB in this region of the rat brain. RESULTS: In the learned helplessness model of depression, induction of CREB in the basolateral amygdala after training decreased the number of escape failures, an antidepressant response. However, expression of CREB before training increased escape failures, and increased immobility in the forced swim test, depressive effects. Expression of CREB in the basolateral amygdala also increased behavioral measures of anxiety in both the open field test and the elevated plus maze, and enhanced cued fear conditioning. CONCLUSIONS: Taken together, these data demonstrate that CREB expression in the basolateral amygdala influences behavior in models of depression, anxiety, and fear. Moreover, in the basolateral amygdala, the temporal expression of CREB in relation to learned helplessness training, determines the qualitative outcome in this animal model of depression.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Proteína Receptora de AMP Cíclico/metabolismo , Depressão/metabolismo , Regulação da Expressão Gênica , Tonsila do Cerebelo/virologia , Animais , Antidepressivos/uso terapêutico , Ansiedade/tratamento farmacológico , Ansiedade/genética , Ansiedade/virologia , Proteínas de Transporte , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Proteína Receptora de AMP Cíclico/genética , Proteína Receptora de AMP Cíclico/fisiologia , Depressão/tratamento farmacológico , Depressão/genética , Depressão/virologia , Modelos Animais de Doenças , Reação de Fuga/efeitos dos fármacos , Reação de Fuga/fisiologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Medo/psicologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Desamparo Aprendido , Imuno-Histoquímica/métodos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Ratos , Ratos Sprague-Dawley , Simplexvirus/metabolismo , Natação/psicologia , Fatores de Tempo
20.
Brain Res ; 968(1): 89-101, 2003 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-12644267

RESUMO

Previously, we have shown that rats administered MDMA from postnatal (P) days 11-20 had reductions in body weight during the period of treatment and as adults they had deficits in sequential and spatial learning and memory. In the present study, to control for weight reductions, we used litters with double the number of offspring to induce growth restriction comparable to that of standard size litters treated with MDMA. Litters were treated twice daily from P11 to 20 with vehicle or MDMA (20 mg/kg) or only weighed. Males, but not females, exposed to MDMA had longer latencies and more errors in the Cincinnati water maze compared to males of the other treatments. In the Morris water maze (210 cm pool, 10x10 cm platform), the MDMA animals were impaired relative to all other treatments during acquisition. Only the MDMA females showed deficits when the platform was shifted to a new location, however, both MDMA males and females were impaired when the location of the platform was again shifted and a reduced platform (5x5 cm) used. No differences were observed in the ability to swim a straight channel, locate a platform with a cue, or the endocrine response to forced swim among the treatment groups. No differences were seen between animals injected with saline and those only weighed. The data suggest that factors, such as growth retardation, multiple injections, or the composition of the litter, do not affect the development of learning and memory impairments resulting from P11 to 20 MDMA exposure. The large litter approach offers a novel method to control for undernutrition during the preweaning period in rodents.


Assuntos
3,4-Metilenodioxianfetamina/análogos & derivados , 3,4-Metilenodioxianfetamina/efeitos adversos , Agonistas de Aminoácidos Excitatórios/efeitos adversos , Deficiências da Aprendizagem/etiologia , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Aprendizagem Seriada/efeitos dos fármacos , Comportamento Espacial/efeitos dos fármacos , 3,4-Metilenodioxianfetamina/administração & dosagem , Hormônio Adrenocorticotrópico/sangue , Envelhecimento , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Comportamento Animal , Constituição Corporal , Peso Corporal , Estudos de Casos e Controles , Corticosterona/sangue , Sinais (Psicologia) , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Feminino , Tamanho da Ninhada de Vivíparos/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Paridade , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Tempo de Reação , Fatores Sexuais , Estresse Fisiológico , Natação/fisiologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA