Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Neuropharmacology ; 240: 109707, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37673332

RESUMO

Sensory networks naturally entrain to rhythmic stimuli like a click train delivered at a particular frequency. Such synchronization is integral to information processing, can be measured by electroencephalography (EEG) and is an accessible index of neural network function. Click trains evoke neural entrainment not only at the driving frequency (F), referred to as the auditory steady state response (ASSR), but also at its higher multiples called the steady state harmonic response (SSHR). Since harmonics play an important and non-redundant role in acoustic information processing, we hypothesized that SSHR may differ from ASSR in presentation and pharmacological sensitivity. In female SD rats, a 2 s-long train stimulus was used to evoke ASSR at 20 Hz and its SSHR at 40, 60 and 80 Hz, recorded from a prefrontal epidural electrode. Narrow band evoked responses were evident at all frequencies; signal power was strongest at 20 Hz while phase synchrony was strongest at 80 Hz. SSHR at 40 Hz took the longest time (∼180 ms from stimulus onset) to establish synchrony. The NMDA antagonist MK801 (0.025-0.1 mg/kg) did not consistently affect 20 Hz ASSR phase synchrony but robustly and dose-dependently attenuated synchrony of all SSHR. Evoked power was attenuated by MK801 at 20 Hz ASSR and 40 Hz SSHR only. Thus, presentation as well as pharmacological sensitivity distinguished SSHR from ASSR, making them non-redundant markers of cortical network function. SSHR is a novel and promising translational biomarker of cortical oscillatory dynamics that may have important applications in CNS drug development and personalized medicine.

2.
Schizophr Res ; 215: 300-307, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31744751

RESUMO

Cognitive functioning in schizophrenia is characterized by a generalized impairment in current cognitive ability based on traditional population-based norms. However, these norms assume a normal cognitive trajectory and do not directly account for illness-related declines from expected cognitive potential. Indeed, schizophrenia patients exhibit even greater deviation between their observed and expected cognitive functioning based on expanded norms that leverage premorbid variables resistant to illness-related features. The current study further quantified the extent to which illness-related features account for this deviation from expectation and assessed its relationship to neurophysiologic (mismatch negativity, P3a, theta oscillations), clinical, and psychosocial functioning in schizophrenia patients. Expected cognitive ability (PENN-CNB global cognition) in patients (n = 684) was calculated using healthy comparison subject (n = 660) weighted regression based on premorbid variables resistant to illness-related decline (demographics, single-word reading, parental education). The magnitude of any deviation between current (observed) and regression-predicted (expected) cognitive ability was calculated. Results indicated that 24% (n = 164) of the total patient population exhibited significant (≥-1.96 SD) deviation between observed and expected global cognitive ability. Interestingly, 20% of the total patient population (n = 136) had "normal" range cognitive performance when using traditional population-based norms, but also had significant deviation from expected cognitive ability. The magnitude of this deviation was associated with more severe neurophysiologic abnormalities, longer illness duration, higher levels of negative symptoms, and worse psychosocial functioning. Assessment of cognitive deviation is thus a complementary metric for characterizing the severity of illness-related cognitive declines in patients, while also reflecting the expression and severity of key endophenotypes of schizophrenia.


Assuntos
Aptidão/fisiologia , Disfunção Cognitiva , Potenciais Evocados/fisiologia , Funcionamento Psicossocial , Esquizofrenia , Ritmo Teta/fisiologia , Adulto , Disfunção Cognitiva/diagnóstico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Esquizofrenia/complicações , Esquizofrenia/diagnóstico , Esquizofrenia/fisiopatologia
3.
Int J Psychophysiol ; 145: 23-29, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30586570

RESUMO

Abnormalities in early auditory information processing (EAIP) contribute to higher-order deficits in cognition and psychosocial functioning in schizophrenia. A passive auditory oddball paradigm is commonly used to evoke event-related potential (ERP) measures of EAIP reflecting auditory sensory registration and deviance detection, including mismatch negativity (MMN) and P3a responses. MMN and P3a have been extensively studied in healthy subjects and neuropsychiatric patient populations and are increasingly used as translational biomarkers in the development of novel therapeutics. Despite widespread use, relatively few studies have examined the constituent oscillatory elements and the extent to which sensory registration and deviance detection represent distinct or intercorrelated processes. This study aimed to determine the factor structure and clinical correlates of these oscillatory measures in schizophrenia patients (n = 706) and healthy comparison subjects (n = 615) who underwent clinical, cognitive, and functional characterization and EEG testing via their participation in the Consortium of Genomics in Schizophrenia (COGS-2) study. Results revealed significant deficits in theta-band (4-7 Hz) evoked power and phase locking in patients. Exploratory factor analyses of both ERP and oscillatory measures revealed two dissociable factors reflecting sensory registration and deviance detection. While each factor shared a significant correlation with social cognition, the deviance detection factor had a unique relationship to multiple cognitive and clinical domains. Results support the continued advancement of functionally relevant oscillatory measures underlying EAIP in the development of precognitive therapeutics.


Assuntos
Encéfalo/fisiopatologia , Cognição/fisiologia , Potenciais Evocados/fisiologia , Esquizofrenia/fisiopatologia , Estimulação Acústica , Adulto , Eletroencefalografia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Tempo de Reação/fisiologia
4.
Mol Psychiatry ; 23(8): 1773-1786, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28948974

RESUMO

Nonsense-mediated RNA decay (NMD) is a highly conserved and selective RNA degradation pathway that acts on RNAs terminating their reading frames in specific contexts. NMD is regulated in a tissue-specific and developmentally controlled manner, raising the possibility that it influences developmental events. Indeed, loss or depletion of NMD factors have been shown to disrupt developmental events in organisms spanning the phylogenetic scale. In humans, mutations in the NMD factor gene, UPF3B, cause intellectual disability (ID) and are strongly associated with autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD) and schizophrenia (SCZ). Here, we report the generation and characterization of mice harboring a null Upf3b allele. These Upf3b-null mice exhibit deficits in fear-conditioned learning, but not spatial learning. Upf3b-null mice also have a profound defect in prepulse inhibition (PPI), a measure of sensorimotor gating commonly deficient in individuals with SCZ and other brain disorders. Consistent with both their PPI and learning defects, cortical pyramidal neurons from Upf3b-null mice display deficient dendritic spine maturation in vivo. In addition, neural stem cells from Upf3b-null mice have impaired ability to undergo differentiation and require prolonged culture to give rise to functional neurons with electrical activity. RNA sequencing (RNAseq) analysis of the frontal cortex identified UPF3B-regulated RNAs, including direct NMD target transcripts encoding proteins with known functions in neural differentiation, maturation and disease. We suggest Upf3b-null mice serve as a novel model system to decipher cellular and molecular defects underlying ID and neurodevelopmental disorders.


Assuntos
Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Deficiências da Aprendizagem/metabolismo , Neurogênese/fisiologia , Inibição Pré-Pulso/fisiologia , Proteínas de Ligação a RNA/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/patologia , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Feminino , Deficiências da Aprendizagem/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Transtornos do Neurodesenvolvimento/metabolismo , Transtornos do Neurodesenvolvimento/patologia , Fenótipo , Células Piramidais/metabolismo , Células Piramidais/patologia , Proteínas de Ligação a RNA/genética , Distribuição Aleatória , Transcrição Gênica
5.
Neuroscience ; 168(2): 498-504, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20338222

RESUMO

Sensorimotor gating, as measured by prepulse inhibition (PPI) of startle, is deficient in human beings with schizophrenia and is greatly reduced in rats after bilateral infusion of N-methyl-D-aspartate (NMDA) into the ventral hippocampus (VH). The disruption of PPI by bilateral VH NMDA infusion is blocked by bilateral medial prefrontal cortex (mPFC) lesions, but not by bilateral lesions of the fornix, which is the principal output pathway of the hippocampal formation of the VH. Tract-tracing studies have shown the presence of additional nonfornical pathways by which the VH and neighboring structures of the amygdala may reach forebrain regions that regulate PPI, including the mPFC. To determine whether these nonfornical pathways might mediate forebrain activation after VH NMDA infusion, we examined the effects of bilateral VH NMDA infusion on c-Fos protein expression in the mPFC and nucleus accumbens (NAC) after sham vs. bilateral fornix lesions. Significant increases of c-Fos expression were observed in both the mPFC and NAC after bilateral VH NMDA infusions. Fornix lesions blocked enhanced c-Fos expression in the NAC but not the mPFC after VH NMDA infusion. The results suggest that an intact fornix may be necessary for VH activation of the NAC, but that the VH uses additional nonfornical projections to activate PPI-regulatory circuits within the mPFC.


Assuntos
Fórnice/fisiologia , Hipocampo/fisiologia , Núcleo Accumbens/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Hipocampo/efeitos dos fármacos , Masculino , N-Metilaspartato/farmacologia , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto
6.
Neuroscience ; 165(2): 601-11, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19854244

RESUMO

The neural substrates regulating sensorimotor gating in rodents are studied in order to understand the basis for gating deficits in clinical disorders such as schizophrenia. N-methyl-D-aspartate (NMDA) infusion into the ventral temporal lobe, including caudal parts of the ventral hippocampal region and amygdala, has been shown to disrupt sensorimotor gating in rats, as measured by prepulse inhibition (PPI) of startle. One working model is that reduced PPI after infusion of NMDA into this region is mediated via its efferents to ventral forebrain structures, i.e. medial prefrontal cortex (mPFC) and nucleus accumbens. Yet, PPI-disruptive effects persist after lesions of the precommissural fornix, the principal output pathway of the hippocampal formation. Here, we aimed to characterize non-fornical forebrain projections from this region that might mediate the PPI-disruptive effects of the ventral temporal lobe. Electrolytic lesions of the precommissural fornix in male Sprague-Dawley rats were followed by infusions of fluorogold into the mPFC or by infusions of biotinylated dextan amine into the ventral temporal lobe. Projections from the ventral subiculum and CA1 regions of the ventral hippocampus to the mPFC and accumbens core and shell were interrupted by fornix lesions. Projections to the mPFC and accumbens from other regions of the ventral temporal lobe, particularly the lateral entorhinal cortex and the embedded olfactory and vomeronasal parts of the caudal amygdala, survived fornix lesions. These additional projections coursed rostrally through the amygdala and emerged via the stria terminalis, interstitial nuclei of the posterior limb of the anterior commissure, and the ventral amygdalofugal pathway. PPI-regulatory portions of the ventral temporal lobe innervate the accumbens and mPFC via multiple routes. It remains to be determined which of these non-fornical projections may be responsible for the persistent regulation of PPI after fornix lesions.


Assuntos
Tonsila do Cerebelo/fisiologia , Percepção Auditiva/fisiologia , Hipocampo/fisiologia , Prosencéfalo/fisiologia , Reflexo de Sobressalto/fisiologia , Tonsila do Cerebelo/anatomia & histologia , Animais , Biotina/análogos & derivados , Dextranos , Fórnice/anatomia & histologia , Fórnice/fisiologia , Hipocampo/anatomia & histologia , Masculino , N-Metilaspartato/metabolismo , Vias Neurais/anatomia & histologia , Vias Neurais/fisiologia , Marcadores do Trato Nervoso , Prosencéfalo/anatomia & histologia , Ratos , Ratos Sprague-Dawley
7.
Neuropsychopharmacology ; 34(7): 1854-64, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19242406

RESUMO

Reduced prepulse inhibition (PPI) of startle provides evidence of deficient sensorimotor gating in several disorders, including schizophrenia. The role of NMDA neurotransmission in the regulation of PPI is unclear, due to cross-species differences in the effects of NMDA antagonists on PPI. Recent reports suggest that drug effects on PPI differ in subgroups of normal humans that differ in the levels of baseline PPI or specific personality domains; here, we tested the effects of these variables on the sensitivity of PPI to the NMDA antagonist, memantine. PPI was measured in male Sprague-Dawley rats, after treatment with memantine (0, 10 or 20 mg/kg, s.c.). Baseline PPI was then measured in 37 healthy adult men. Next, subjects were tested twice, in a double-blind crossover design, comparing either (1) placebo vs 20 mg of the NMDA antagonist memantine (n=19) or (2) placebo vs 30 mg memantine (n=18). Tests included measures of acoustic startle amplitude, PPI, autonomic indices and subjective self-rating scales. Memantine had dose- and interval-dependent effects on PPI in rats. Compared with vehicle, 10 mg/kg increased short-interval (10-20 ms) PPI, and 20 mg/kg decreased long-interval (120 ms) PPI. In humans, memantine caused dose-dependent effects on psychological and somatic measures: 20 mg was associated with increased ratings of happiness, and 30 mg was associated with increased ratings of dizziness. PPI at the 120 ms prepulse interval was increased by 20 mg, but not 30 mg of memantine. Subgroups most sensitive to the PPI-enhancing effects of memantine were those with low baseline PPI, or with personality scale scores suggestive of high novelty seeking, high sensation seeking, or high disinhibition. NMDA blockade with memantine appears to have dose- and interval-dependent effects on sensorimotor gating in rats and humans, particularly among specific subgroups of normal human subjects. These findings are discussed as they relate to consistencies across other studies in humans, as well as apparent inconsistencies in the NMDA regulation of PPI across species.


Assuntos
Antagonistas de Aminoácidos Excitatórios/farmacologia , Memantina/farmacologia , Inibição Neural/efeitos dos fármacos , Filtro Sensorial/efeitos dos fármacos , Estimulação Acústica/métodos , Adulto , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Estudos Cross-Over , Relação Dose-Resposta a Droga , Método Duplo-Cego , Eletromiografia , Humanos , Masculino , Personalidade , Psicoacústica , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Reflexo de Sobressalto/efeitos dos fármacos , Inquéritos e Questionários , Fatores de Tempo , Adulto Jovem
8.
Psychopharmacology (Berl) ; 203(4): 723-35, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19066855

RESUMO

RATIONALE: Prepulse inhibition (PPI) is an operational measure of sensorimotor gating that is deficient in schizophrenia patients. In rats, PPI deficits induced by dopamine (DA) agonists are reversed by antipsychotics. Inhibition of the striatum-rich phosphodiesterase (PDE)10A may represent a novel antipsychotic mechanism. Previous studies were controversial, showing antipsychotic-like profiles in measures of PPI for the preferential PDE10A inhibitor papaverine (PAP) but not the novel PDE10A inhibitor TP-10. OBJECTIVE: The aim of the study was to evaluate the antipsychotic profile of PAP in rats using PPI. MATERIALS AND METHODS: PPI deficits were induced in rats by apomorphine (APO; 0.1, 0.5 mg/kg) or D: -amphetamine (AMPH; 4 mg/kg). PAP (3, 10, 30 mg/kg) or haloperidol (HAL; 0.1 mg/kg) was tested against these agonists in Sprague-Dawley (SD) or Wistar (WI) rats. Prepulse intervals ranged from 10 to 120 ms. Further tests evaluated the effects of PAP on spontaneous locomotion, AMPH (1 mg/kg)-induced hyperlocomotion, and core body temperature (T degrees ). RESULTS: HAL reversed APO-induced PPI deficits but PAP failed to reverse APO- and AMPH-induced PPI deficits at all doses, strains, pretreatment times, and prepulse intervals. PAP (30 mg/kg) significantly reduced AMPH hyperlocomotion in SD rats, and a similar pattern was detected in WI rats. This PAP dose also strongly reduced spontaneous locomotion and T degrees in SD rats. CONCLUSION: Our study does not support an antipsychotic-like profile of PAP in dopaminergic PPI models.


Assuntos
Antipsicóticos/farmacologia , Papaverina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/fisiologia , Reflexo de Sobressalto/efeitos dos fármacos , Anfetamina/farmacologia , Animais , Antipsicóticos/farmacocinética , Antipsicóticos/uso terapêutico , Apomorfina/farmacologia , Comportamento Animal/efeitos dos fármacos , Disponibilidade Biológica , Temperatura Corporal/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Haloperidol/farmacologia , Haloperidol/uso terapêutico , Masculino , Atividade Motora/efeitos dos fármacos , Papaverina/farmacocinética , Inibidores de Fosfodiesterase/farmacocinética , Ratos , Ratos Sprague-Dawley , Ratos Wistar
9.
Pharmacol Biochem Behav ; 88(3): 306-11, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17900675

RESUMO

Prepulse inhibition of startle (PPI) is an operational measure of sensorimotor gating that is impaired in schizophrenia and is disrupted in rats by dopamine (DA) agonists like apomorphine (APO). Using acoustic prepulses and acoustic startle pulses, previous studies have demonstrated heritable strain differences between Sprague Dawley (SD) and Long Evans (LE) rats in the sensitivity to the PPI-disruptive effects of APO. As PPI deficits in schizophrenia are evident with both uni- and cross-modal stimuli, we tested whether strain differences in the gating-disruptive effects of APO occur with a cross-modal visual and acoustic stimulus combination. APO caused a dose-dependent disruption of both acoustic and visual PPI in SD rats. Compared to LE rats, SD rats were more sensitive to the PPI-disruptive effects of APO with both acoustic and visual PPI. These findings suggest that SD vs. LE strain differences in PPI APO sensitivity are mediated outside of the auditory system, within higher circuitry that regulates or processes multi-modal information. The present findings provide further validation for this heritable model of impaired sensorimotor gating in schizophrenia, which can be detected across multiple sensory modalities.


Assuntos
Apomorfina/farmacologia , Agonistas de Dopamina/farmacologia , Reflexo de Sobressalto/efeitos dos fármacos , Estimulação Acústica , Análise de Variância , Animais , Luz , Masculino , Atividade Motora/efeitos dos fármacos , Estimulação Luminosa , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Especificidade da Espécie
10.
J Neural Transm (Vienna) ; 114(7): 893-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17594127

RESUMO

Post-mortem studies have provided evidence for abnormalities of the gamma-aminobutyric acid (GABA)-ergic system in schizophrenia. The calcium-binding proteins (CBPs), parvalbumin (PV), calbindin (CB) and calretinin (CR) can be used as markers for specific subpopulations of GABAergic neurons in the brain. Isolation rearing of rats is a non-pharmacological, non-lesion manipulation that leads to deficits in prepulse inhibition of the startle reflex (PPI) and other behavioural and neurochemical alterations reminiscent of schizophrenia. Female rats were reared in social housing (groups of three) or singly for 11 weeks post weaning and PPI was measured. Brains were removed and hippocampal CBP- containing neurons determined following immunocytochemical staining. Compared to socially housed rats, isolated rats exhibited PPI deficits and reductions in PV and CB-immunoreactive cells in the hippocampus, with no significant change in CR. These findings demonstrate selective abnormalities of sub-populations of GABAergic interneurons in the hippocampus of isolation reared rats, which resemble the neuronal deficits seen in this region in schizophrenia.


Assuntos
Hipocampo/metabolismo , Hipocampo/patologia , Abrigo para Animais , Parvalbuminas/deficiência , Proteína G de Ligação ao Cálcio S100/metabolismo , Isolamento Social/psicologia , Animais , Biomarcadores/química , Biomarcadores/metabolismo , Calbindina 2 , Calbindinas , Contagem de Células , Feminino , Interneurônios/química , Interneurônios/metabolismo , Interneurônios/patologia , Parvalbuminas/biossíntese , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/fisiologia , Proteína G de Ligação ao Cálcio S100/biossíntese , Ácido gama-Aminobutírico/fisiologia
11.
Neuroscience ; 135(2): 385-94, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16125865

RESUMO

BACKGROUND: Sensorimotor gating, as measured by prepulse inhibition of the startle reflex, is deficient in schizophrenia patients, and in rats after specific manipulations of limbic cortico-striato-pallido-thalamic circuitry. For example, prepulse inhibition in rats is disrupted after D1 blockade in the medial prefrontal cortex, and after N-methyl-D-aspartate infusion into the ventral hippocampus. In the present study, we examined whether these two substrates form part of an integrated circuit regulating sensorimotor gating, which might contribute to the loss of prepulse inhibition in patient populations. METHODS: Prepulse inhibition was assessed in male Sprague-Dawley rats after systemic or intra-medial prefrontal cortex administration of the D1 antagonist, SCH 23390. Separate rats received intra-medial prefrontal cortex infusion of the retrograde transported label Fluoro-Gold. In rats with sham or electrolytic lesions of the medial prefrontal cortex, prepulse inhibition was tested after infusion of N-methyl-D-aspartate or vehicle into ventral hippocampus regions that were determined to send projections to the medial prefrontal cortex. RESULTS: Prepulse inhibition was disrupted after systemic SCH 23390 treatment and after infusion of SCH 23390 into medial prefrontal cortex sites within the prelimbic and cingulate cortices. Fluoro-Gold infusion into these medial prefrontal cortex sites labeled cells in the ventral hippocampus complex, including regions CA1 and entorhinal cortex. N-methyl-D-aspartate infusions into these ventral hippocampus regions disrupted prepulse inhibition in rats after sham but not electrolytic lesions of the medial prefrontal cortex. CONCLUSIONS: Prepulse inhibition appears to be regulated by interacting substrates within the ventral hippocampus and MPFC. Specifically, NMDA activation of the ventral hippocampus appears to disrupt prepulse inhibition in a manner that is dependent on the integrity of infralimbic or cingulate cortical regions that also support a D1-mediated regulation of prepulse inhibition. Conceivably, dysfunction within these hippocampal-frontal circuits may contribute to sensorimotor gating deficits in schizophrenia.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/efeitos dos fármacos , N-Metilaspartato/farmacologia , Córtex Pré-Frontal/fisiologia , Receptores de Dopamina D1/fisiologia , Reflexo de Sobressalto/fisiologia , Estimulação Acústica/métodos , Análise de Variância , Animais , Comportamento Animal , Benzazepinas/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Hipocampo/fisiologia , Masculino , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/efeitos dos fármacos , Estilbamidinas/metabolismo
12.
Neuroscience ; 123(3): 675-85, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14706779

RESUMO

Prepulse inhibition (PPI) of startle is a measure of sensorimotor gating that is impaired in schizophrenia. We have reported that PPI is regulated by the ventral hippocampus (VH) and that the PPI disruptive effects of the dopamine agonist apomorphine are enhanced 4 weeks after excitotoxic lesions of the VH. The mechanisms responsible for the VH influence on PPI are not understood, but have been ascribed to interactions between the VH and nucleus accumbens. In the present study, we examined whether the VH influence on PPI and its dopaminergic regulation is dependent on the integrity of the VH-accumbens projection via the fornix. First, the PPI-disruptive effects of intra-VH NMDA infusion were assessed after sham or electrolytic transection of the fornix. Second, the PPI-disruptive effects of apomorphine were assessed 1 month after excitotoxic or electrolytic lesions of the VH, or after fornix transection. Intra-VH N-methyl-D-aspartate infusion significantly disrupted PPI; this effect was unaffected by fornix lesions. The PPI-disruptive effects of apomorphine were significantly enhanced by excitotoxic or electrolytic lesions of the VH, but not by fornix transection. The influence of the VH on PPI and its dopaminergic regulation does not appear to be mediated via the fornix. The enhanced sensitivity to the PPI-disruptive effects of apomorphine after VH lesions is not dependent on excitotoxin-induced changes in the VH or its downstream projections.


Assuntos
Apomorfina/farmacologia , Fórnice/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Reflexo de Sobressalto/efeitos dos fármacos , Animais , Fórnice/fisiologia , Hipocampo/fisiologia , Inibição Neural/fisiologia , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/fisiologia
13.
Neuroscience ; 119(1): 233-40, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12763084

RESUMO

Rearing rats in social isolation from weaning into adulthood leads to deficits in prepulse inhibition and alterations in monoamine systems that modulate prepulse inhibition. For example, rats reared in social isolation have elevated dopamine levels in the nucleus accumbens. Previous studies in rats have shown that nucleus accumbens dopamine depletion with 6-hydroxydopamine blocks the prepulse inhibition-disruptive effects of amphetamine, an indirect dopamine agonist. We tested the hypothesis that prepulse-inhibition deficits in isolation-reared rats are dependent on elevated dopamine levels in the nucleus accumbens. Specifically, we examined whether nucleus accumbens dopamine depletion would attenuate the isolation-induced disruption of prepulse inhibition. Isolation-housed female Long-Evans rats exhibited deficient prepulse inhibition. At 9 weeks post weaning, bilateral injections of 6-hydroxydopamine (8 microg/side) or ascorbic acid vehicle (0.1%) into the nucleus accumbens of social and isolation-reared rats were performed (8-10 rats per group). One week after surgery, prepulse inhibition deficits were exhibited by isolation-reared rats that received vehicle infusion into the nucleus accumbens, but not by those that received 6-hydroxydopamine infusions into the nucleus accumbens. 6-Hydroxydopamine infusions did not significantly change prepulse inhibition in socially reared rats. Behavioral and neurochemical evidence of nucleus accumbens dopamine depletion included: 1) a blockade of amphetamine-stimulated locomotor activity in nucleus accumbens 6-hydroxydopamine-infused isolated and socially reared rats; and 2) high performance liquid chromatography measurements demonstrating a significant depletion of accumbens dopamine and its major metabolites, in addition to decreases in dopamine, homovanillic acid, and 3,4-dihydroxyphenylacetic acid levels in the frontal cortex and anterior caudate. These data indicate that dopamine in the nucleus accumbens plays an essential role in the prepulse inhibition deficits associated with isolation rearing in female Long-Evans rats. The implication of a central role of nucleus accumbens dopamine in prepulse inhibition deficits in an animal model provides further evidence for a link between overactive dopamine function and sensorimotor-gating deficits in patients with schizophrenia.


Assuntos
Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Isolamento Social/psicologia , Estimulação Acústica , Adrenérgicos/toxicidade , Anfetamina/farmacologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Cromatografia Líquida de Alta Pressão/métodos , Feminino , Atividade Motora/fisiologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Núcleo Accumbens/lesões , Oxidopamina/toxicidade , Ratos , Ratos Long-Evans , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/fisiologia
14.
Psychopharmacology (Berl) ; 158(3): 230-40, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11713612

RESUMO

RATIONALE: Prepulse inhibition (PPI), a cross-species measure of sensorimotor gating, is impaired in certain neuropsychiatric disorders, including schizophrenia. This study was designed to assess the effects of the D2-family agonist pergolide in rats, in anticipation of human studies of the dopaminergic regulation of PPI. METHODS: The effects of pergolide (0.0001-0.5 mg/kg) on PPI of the acoustic startle reflex were studied in rats using a wide range of prepulse intensities [1-15 dB(A) over background] and prepulse intervals (5-100 ms, onset to onset). Studies also examined the effects of the D2 antagonist haloperidol on pergolide-induced changes in PPI. RESULTS: Pergolide exhibited dose- and stimulus-dependent effects on PPI. Pergolide increased PPI when startle stimuli were preceded by weak prepulses [1-5 dB(A) over background] at the longest prepulse interval (100 ms), or intense prepulses [15 dB(A) over background] at short prepulse intervals (5-20 ms). Pergolide (0.5 mg/kg) also decreased PPI elicited by intense prepulses at long intervals (60-100 ms). Both PPI-enhancing and PPI-disruptive effects of pergolide were reversed by the D2 antagonist haloperidol. CONCLUSIONS: These effects of pergolide suggest that D2 substrates mediate opposing influences on PPI under different stimulus conditions. The dopaminergic regulation of sensorimotor gating appears to interact with stimulus characteristics such as relative intensity and temporal separation, allowing for dynamic shifts in both the quantity and quality of "gated" information.


Assuntos
Agonistas de Dopamina/farmacologia , Pergolida/farmacologia , Desempenho Psicomotor/efeitos dos fármacos , Reflexo de Sobressalto/efeitos dos fármacos , Animais , Antagonistas dos Receptores de Dopamina D2 , Relação Dose-Resposta a Droga , Masculino , Desempenho Psicomotor/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/fisiologia , Reflexo de Sobressalto/fisiologia
15.
Pharmacol Biochem Behav ; 70(2-3): 219-26, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11701191

RESUMO

Prepulse inhibition (PPI), a measure of sensorimotor gating, is reduced in schizophrenia patients and in rats treated with dopamine (DA) agonists. Reported strain and supplier-based differences in sensitivity to PPI-disruptive effects of DA agonists presumably reflect the differential impact of genetics and/or environment on DAergic substrates regulating PPI. In 2000, Harlan Laboratories established a Texas Sprague-Dawley line (SDHt; facility 211) using breeders from Indianapolis (SDHi; facility 202A). SDHi rats had been used, approximately 11 years earlier, to establish a colony in San Diego (SDHsd; facility 235). SDHt and SDHi rats are thus genetically similar, but raised in distinct environments; approximately 11 years of genetic "drift" separates SDHsd rats from both SDHi and SDHt rats. Harlan Long-Evans hooded rats (LEH; Madison, WI; facility 207) are genetically distinct from albino SDH. All except SDHsd rats were shipped to our facility by air freight. We used SDHt, SDHi, SDHsd, and LEH rats to assess genetic and environmental contributions to the DAergic regulation of PPI. Acoustic startle/PPI were assessed in rats treated with the D1/D2 agonist apomorphine (APO), the D2 agonist quinpirole, or the D1 agonist SKF 82958. The relative sensitivities to the PPI-disruptive effects were: APO: SDHt=SDHsd=SDHi>>LEH; SKF 82958: SDHt=SDHsd=SDHi (LEH not sensitive); quinpirole: SDHt=SDHsd=SDHi; SDHi>LEH. Strain/supplier differences in sensitivity to drug effects on startle magnitude did not correspond to patterns of PPI sensitivity. In these rats, strain differences in the DAergic regulation of PPI are most easily explained by genetic, rather than environmental influences that differentially impact both D1 and D2 substrates. This finding is consistent with published reports in other strains. Pharmacogenetic studies of PPI in rats may identify a genetic basis for a model of deficient sensorimotor gating in schizophrenia.


Assuntos
Agonistas de Dopamina/farmacologia , Meio Ambiente , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/genética , Animais , Apomorfina/farmacologia , Dopamina/genética , Masculino , Fenótipo , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Especificidade da Espécie
16.
Biol Psychiatry ; 50(8): 578-85, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11690592

RESUMO

BACKGROUND: Functional magnetic resonance imaging (fMRI) studies in neuropsychiatric populations will be enhanced by "on-line" tasks that assess brain activation linked to neurocognitive and psychophysiological functions. In some cases, task modifications may be required for use in an fMRI environment. Prepulse inhibition (PPI) of the startle reflex is an operational measure of sensorimotor gating that is deficient in specific neuropsychiatric disorders, including schizophrenia, Huntington's disease, and Tourette's syndrome (TS). This study examined whether a modified "fMRI-friendly" PPI paradigm is suitable for use in children and adequately sensitive to detect PPI deficits in TS. METHODS: Bilateral eyeblink PPI was measured in children using chin air puffs to elicit startle and prepuffs to the dorsal hand surface as inhibiting stimuli. This paradigm involved no metallic objects or acoustic stimuli, making it suitable for an fMRI environment that is magnetically sensitive and acoustically complex. Children were also assessed in a "standard" acoustic PPI paradigm. RESULTS: Robust startle was elicited via either puffs or noise bursts, and these responses were inhibited by prepuffs and prepulses, respectively. Compared to control subjects, children with TS exhibited comparable startle magnitude and habituation but significantly reduced prepuff inhibition and acoustic PPI. CONCLUSIONS: Sensorimotor gating can be assessed in an "fMRI-friendly" paradigm that detects inhibitory deficits in TS.


Assuntos
Piscadela/fisiologia , Imageamento por Ressonância Magnética , Inibição Neural/fisiologia , Tato/fisiologia , Síndrome de Tourette/fisiopatologia , Estimulação Acústica , Adolescente , Encéfalo/fisiopatologia , Mapeamento Encefálico , Criança , Feminino , Humanos , Masculino , Estimulação Física , Síndrome de Tourette/diagnóstico
17.
Psychopharmacology (Berl) ; 156(2-3): 117-54, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11549216

RESUMO

RATIONALE: Patients with schizophrenia exhibit deficits in an operational measure of sensorimotor gating: prepulse inhibition (PPI) of startle. Similar deficits in PPI are produced in rats by pharmacological or developmental manipulations. These experimentally induced PPI deficits in rats are clearly not animal models of schizophrenia per se, but appear to provide models of sensorimotor gating deficits in schizophrenia patients that have face, predictive, and construct validity. In rodents, disruptions in PPI of startle are produced by: stimulation of D2 dopamine (DA) receptors, produced by amphetamine or apomorphine; activation of serotonergic systems, produced by serotonin (5-HT) releasers or direct agonists at multiple serotonin receptors; and blockade of N-methyl-D-aspartate (NMDA) receptors, produced by drugs such as phencyclidine (PCP). Accordingly, dopaminergic, serotonergic, and glutamatergic models of disrupted PPI have evolved and have been applied to the identification of potential antipsychotic treatments. In addition, some developmental manipulations, such as isolation rearing, have provided non-pharmacological animal models of the PPI deficits seen in schizophrenia. OBJECTIVE: This review summarizes and evaluates studies assessing the effects of systemic drug administrations on PPI in rats. METHODS: Studies examining systemic drug effects on PPI in rats prior to January 15, 2001 were compiled and organized into six annotated appendices. Based on this catalog of studies, the specific advantages and disadvantages of each of the four main PPI models used in the study of antipsychotic drugs were critically evaluated. RESULTS: Despite some notable inconsistencies, the literature provides strong support for significant disruptions in PPI in rats produced by DA agonists, 5-HT2 agonists, NMDA antagonists, and isolation rearing. Each of these models exhibits sensitivity to at least some antipsychotic medications. While the PPI model based on the effects of direct DA agonists is the most well-validated for the identification of known antipsychotics, the isolation rearing model also appears to be sensitive to both typical and atypical antipsychotics. The 5-HT PPI model is less generally sensitive to antipsychotic medications, but can provide insight into the contribution of serotonergic systems to the actions of newer antipsychotics that act upon multiple receptors. The deficits in PPI produced by NMDA antagonists appear to be more sensitive to clozapine-like atypical antipsychotics than to typical antipsychotics. Hence, despite some exceptions to this generalization, the NMDA PPI model might aid in the identification of novel or atypical antipsychotic medications. CONCLUSIONS: Studies of drug effects on PPI in rats have generated four distinctive models that have utility in the identification of antipsychotic medications. Because each of these models has specific advantages and disadvantages, the choice of model to be used depends upon the question being addressed. This review should help to guide such decisions.


Assuntos
Reflexo de Sobressalto/efeitos dos fármacos , Psicologia do Esquizofrênico , Animais , Humanos , Esquizofrenia/tratamento farmacológico
18.
Psychopharmacology (Berl) ; 156(2-3): 194-215, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11549223

RESUMO

RATIONALE: Sensorimotor gating of the startle reflex can be assessed across species, using similar stimuli to elicit similar responses. Prepulse inhibition (PPI), a measure of sensorimotor gating, is reduced in patients with some neuropsychiatric disorders, and in rats after manipulations of limbic cortex, striatum, pallidum or pontine tegmentum ("CSPP" circuitry). OBJECTIVE: To review the current knowledge of the neural circuit regulation of PPI in rats, and to anticipate the future challenges facing this line of inquiry. METHODS: The published literature was reviewed and critically evaluated. RESULTS: Limbic CSPP circuitry has been studied in rats to reveal the neurochemical and neuroanatomical substrates regulating PPI at a high level of resolution. In translational cross-species research, this detailed circuit information is used as a "blueprint" to identify substrates that may lead to PPI deficits in psychiatrically disordered humans. Some human disorders with identifiable, localized lesions in CSPP circuitry may provide direct validation for the contribution of CSPP circuitry to this cross-species model. The rapid collection of experimental data supporting this cross-species PPI circuit "blueprint" has supported continuing advances in the development of theoretical models for understanding how this circuitry normally functions to regulate PPI. Such models are needed for building a conceptual framework for understanding the role of this circuitry in the regulation of sensorimotor gating in normal humans, and in the relative loss of sensorimotor gating, and the resulting clinical consequences, in individuals with particular neuropsychiatric disorders. CONCLUSIONS: Our understanding of the neural regulation of PPI has increased tremendously over the past 15 years. Progress has come in "broad strokes", and a number of important details and complex questions remain to be addressed. It is anticipated that this is a "work in progress", and that the precise models for the neural regulation of PPI will evolve substantially in the coming years.


Assuntos
Rede Nervosa/fisiologia , Reflexo de Sobressalto/fisiologia , Animais , Encéfalo/fisiologia , Ratos
19.
Psychopharmacology (Berl) ; 156(2-3): 234-58, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11549226

RESUMO

RATIONALE: Since the mid-1970s, cross-species translational studies of prepulse inhibition (PPI) have increased at an astounding pace as the value of this neurobiologically informative measure has been optimized. PPI occurs when a relatively weak sensory event (the prepulse) is presented 30-500 ms before a strong startle-inducing stimulus, and reduces the magnitude of the startle response. In humans, PPI occurs in a robust, predictable manner when the prepulse and startling stimuli occur in either the same or different modalities (acoustic, visual, or cutaneous). OBJECTIVE: This review covers three areas of interest in human PPI studies. First, we review the normal influences on PPI related to the underlying construct of sensori- (prepulse) motor (startle reflex) gating. Second, we review PPI studies in psychopathological disorders that form a family of gating disorders. Third, we review the relatively limited but interesting and rapidly expanding literature on pharmacological influences on PPI in humans. METHODS: All studies identified by a computerized literature search that addressed the three topics of this review were compiled and evaluated. The principal studies were summarized in appropriate tables. RESULTS: The major influences on PPI as a measure of sensorimotor gating can be grouped into 11 domains. Most of these domains are similar across species, supporting the value of PPI studies in translational comparisons across species. The most prominent literature describing deficits in PPI in psychiatrically defined groups features schizophrenia-spectrum patients and their clinically unaffected relatives. These findings support the use of PPI as an endophenotype in genetic studies. Additional groups of psychopathologically disordered patients with neuropathology involving cortico-striato-pallido-pontine circuits exhibit poor gating of motor, sensory, or cognitive information and corresponding PPI deficits. These groups include patients with obsessive compulsive disorder, Tourette's syndrome, blepharospasm, temporal lobe epilepsy with psychosis, enuresis, and perhaps posttraumatic stress disorder (PTSD). Several pharmacological manipulations have been examined for their effects on PPI in healthy human subjects. In some cases, the alterations in PPI produced by these drugs in animals correspond to similar effects in humans. Specifically, dopamine agonists disrupt and nicotine increases PPI in at least some human studies. With some other compounds, however, the effects seen in humans appear to differ from those reported in animals. For example, the PPI-increasing effects of the glutamate antagonist ketamine and the serotonin releaser MDMA in humans are opposite to the PPI-disruptive effects of these compounds in rodents. CONCLUSIONS: Considerable evidence supports a high degree of homology between measures of PPI in rodents and humans, consistent with the use of PPI as a cross-species measure of sensorimotor gating. Multiple investigations of PPI using a variety of methods and parameters confirm that deficits in PPI are evident in schizophrenia-spectrum patients and in certain other disorders in which gating mechanisms are disturbed. In contrast to the extensive literature on clinical populations, much more work is required to clarify the degree of correspondence between pharmacological effects on PPI in healthy humans and those reported in animals.


Assuntos
Transtornos Mentais/psicologia , Psicotrópicos/farmacologia , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/fisiologia , Humanos , Transtornos Mentais/fisiopatologia , Psicologia do Esquizofrênico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...