Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Pharmacol Drug Dev ; 12(6): 625-638, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37191222

RESUMO

ESB1609 is a small-molecule sphingosine-1-phosphate-5 receptor-selective agonist designed to restore lipid homeostasis by promoting cytosolic egress of sphingosine-1-phosphate to reduce abnormal levels of ceramide and cholesterol in disease. A phase 1 study was conducted in healthy volunteers to determine the safety, tolerability, and pharmacokinetics of ESB1609. Following single oral doses, ESB1609 demonstrated linear pharmacokinetics in plasma and cerebrospinal fluid (CSF) for formulations containing sodium laurel sulfate. Plasma and CSF median time to maximum drug concentration (tmax ) were reached by 4-5 hours and 6-10 hours, respectively. The delay in achieving tmax in CSF relative to plasma, likely due to the high protein binding of ESB1609, was also observed in 2 rat studies. Continuous CSF collection via indwelling catheters confirmed that a highly protein-bound compound is measurable and established the kinetics of ESB1609 in human CSF. Mean plasma terminal elimination half-lives ranged from 20.2 to 26.8 hours. The effect of either a high-fat or standard meal increased maximum plasma concentration and area under the concentration-time curve from time 0 to infinity compared to the fasted state by 2.42-4.34-fold higher, but tmax and half-life remained the same irrespective of fed state. ESB1609 crosses the blood-brain barrier with CSF:plasma ratios ranging between 0.04% and 0.07% across dose levels. ESB1609 demonstrated a favorable safety and tolerability profile at exposures expected to be efficacious.


Assuntos
Jejum , Humanos , Animais , Ratos , Receptores de Esfingosina-1-Fosfato , Administração Oral , Área Sob a Curva
2.
Neurol Ther ; 11(3): 1353-1374, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35779189

RESUMO

INTRODUCTION: This randomized, double-blind, placebo-controlled study in healthy volunteers assessed the safety, tolerability, and pharmacokinetics of single ascending doses of intravenously administered NX210-a linear peptide derived from subcommissural organ-spondin-and explored the effects on blood/urine biomarkers and cerebral activity. METHODS: Participants in five cohorts (n = 8 each) were randomized to receive a single intravenous dose of NX210 (n = 6 each) (0.4, 1.25, 2.5, 5, and 10 mg/kg) or placebo (n = 2 each); in total, 10 and 29 participants received placebo and NX210, respectively. Blood samples were collected for pharmacokinetics within 180 min post dosing. Plasma and urine were collected from participants (cohorts: 2.5, 5, and 10 mg/kg) for biomarker analysis and electroencephalography (EEG) recordings within 48 h post dosing. Safety/tolerability and pharmacokinetic data were assessed before ascending to the next dose. RESULTS: The study included 39 participants. All dosages were safe and well tolerated. All treatment-emergent adverse events (n = 17) were of mild severity and resolved spontaneously (except one with unknown outcome). Twelve treatment-emergent adverse events (70.6%) were deemed drug related; seven of those (58.3%) concerned nervous system disorders (dizziness, headache, and somnolence). The pharmacokinetic analysis indicated a short half-life in plasma (6-20 min), high apparent volume of distribution (1870-4120 L), and rapid clearance (7440-16,400 L/h). In plasma, tryptophan and homocysteine showed dose-related increase and decrease, respectively. No drug dose effect was found for the glutamate or glutamine plasma biomarkers. Nevertheless, decreased blood glutamate and increased glutamine were observed in participants treated with NX210 versus placebo. EEG showed a statistically significant decrease in beta and gamma bands and a dose-dependent increasing trend in alpha bands. Pharmacodynamics effects were sustained for several hours (plasma) or 48 h (urine and EEG). CONCLUSION: NX210 is safe and well tolerated and may exert beneficial effects on the central nervous system, particularly in terms of cognitive processing.

3.
Anesthesiology ; 127(1): 20-35, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28459733

RESUMO

BACKGROUND: Cyclopropyl-methoxycarbonylmetomidate (ABP-700) is a new "soft" etomidate analog. The primary objectives of this first-in-human study were to describe the safety and efficacy of ABP-700 and to determine its maximum tolerated dose. Secondary objectives were to characterize the pharmacokinetics of ABP-700 and its primary metabolite (cyclopropyl-methoxycarbonyl acid), to assess the clinical effects of ABP-700, and to investigate the dose-response and pharmacokinetic/pharmacodynamic relationships. METHODS: Sixty subjects were divided into 10 cohorts and received an increasing, single bolus of either ABP-700 or placebo. Safety was assessed by clinical laboratory evaluations, infusion-site reactions, continuous monitoring of vital signs, physical examination, adverse event monitoring, and adrenocorticotropic hormone stimulation testing. Clinical effects were assessed with modified observer's assessment of alertness/sedation and Bispectral Index monitoring. Pharmacokinetic parameters were calculated. RESULTS: Stopping criteria were met at 1.00 mg/kg dose. No serious adverse events were reported. Adverse events were dose-dependent and comprised involuntary muscle movement, tachycardia, and ventilatory effects. Adrenocorticotropic hormone stimulation evoked a physiologic cortisol response in all subjects, no different from placebo. Pharmacokinetics were dose-proportional. A three-compartment pharmacokinetic model described the data well. A rapid onset of anesthesia/sedation after bolus administration and also a rapid recovery were observed. A quantitative concentration-effect relationship was described for the modified observer's assessment of alertness/sedation and Bispectral Index. CONCLUSIONS: This first-in-human study of ABP-700 shows that ABP-700 was safe and well tolerated after single-bolus injections up to 1.00 mg/kg. Bolus doses of 0.25 and 0.35 mg/kg were found to provide the most beneficial clinical effect versus side-effect profile.


Assuntos
Etomidato/análogos & derivados , Etomidato/farmacologia , Hipnóticos e Sedativos/farmacologia , Adolescente , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Etomidato/administração & dosagem , Feminino , Humanos , Hipnóticos e Sedativos/administração & dosagem , Infusões Intravenosas , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Valores de Referência , Adulto Jovem
4.
Science ; 314(5796): 130-3, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-17023659

RESUMO

Ubiquitin-positive, tau- and alpha-synuclein-negative inclusions are hallmarks of frontotemporal lobar degeneration with ubiquitin-positive inclusions and amyotrophic lateral sclerosis. Although the identity of the ubiquitinated protein specific to either disorder was unknown, we showed that TDP-43 is the major disease protein in both disorders. Pathologic TDP-43 was hyper-phosphorylated, ubiquitinated, and cleaved to generate C-terminal fragments and was recovered only from affected central nervous system regions, including hippocampus, neocortex, and spinal cord. TDP-43 represents the common pathologic substrate linking these neurodegenerative disorders.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Química Encefálica , Proteínas de Ligação a DNA/análise , Demência/metabolismo , Medula Espinal/química , Ubiquitina/análise , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/patologia , Anticorpos Monoclonais , Córtex Cerebral/química , Córtex Cerebral/patologia , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Demência/genética , Demência/patologia , Imunofluorescência , Hipocampo/química , Hipocampo/patologia , Humanos , Immunoblotting , Dados de Sequência Molecular , Neurônios Motores/química , Neurônios Motores/patologia , Neurônios/química , Neurônios/patologia , Fragmentos de Peptídeos/química , Fosforilação , Medula Espinal/patologia
5.
Am J Pathol ; 169(4): 1343-52, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17003490

RESUMO

Frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U) is a common neuropathological subtype of frontotemporal dementia. Although this subtype of frontotemporal dementia is defined by the presence of ubiquitin-positive but tau- and alpha-synuclein-negative inclusions, it is unclear whether all cases of FTLD-U have the same underlying pathogenesis. Examination of tissue sections from FTLD-U brains stained with anti-ubiquitin antibodies revealed heterogeneity in the morphological characteristics of pathological inclusions among subsets of cases. Three types of FTLD-U were delineated based on morphology and distribution of ubiquitin-positive inclusions. To address the hypothesis that FTLD-U is pathologically heterogeneous, novel monoclonal antibodies (mAbs) were generated by immunization of mice with high molecular mass (Mr > 250 kd) insoluble material prepared by biochemical fractionation of FTLD-U brains. Novel mAbs were identified that immunolabeled all of the ubiquitin-positive inclusions in one subset of FTLD-U cases, whereas other mAbs stained the ubiquitin-positive inclusions in a second subset of cases. These novel mAbs did not stain inclusions in other neurodegenerative disorders, including tauopathies and alpha-synucleinopathies. Therefore, ubiquitin immunohistochemistry and the immunostaining properties of the novel mAbs generated here suggest that FTLD-U is pathologically heterogeneous. Identification of the disease proteins recognized by these mAbs will further advance understanding of molecular substrates of FTLD-U neurodegenerative pathways.


Assuntos
Anticorpos Monoclonais/imunologia , Demência/patologia , Lobo Frontal/patologia , Corpos de Inclusão/química , Lobo Temporal/patologia , Ubiquitina/análise , Idoso , Animais , Anticorpos Monoclonais/biossíntese , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Pessoa de Meia-Idade , Ubiquitina/imunologia
6.
Clin Ther ; 27(10): 1596-606, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16330295

RESUMO

BACKGROUND: The high prevalence rates of both Alzheimer's disease (AD) and type 2 diabetes mellitus in the elderly population suggest that concomitant pharmacotherapy is likely. Given the renal tubular transport and extensive urinary excretion of memantine and metformin, it was of interest to assess the pharmacokinetic and pharmacodynamic interaction with glyburide/metformin. OBJECTIVE: The primary goal of this study was to determine whether an in vivo pharmacokinetic or pharmacodynamic interaction exists between memantine (an uncompetitive, moderate-affinity, N-methyl-D-aspartate receptor antagonist with fast blocking/unblocking kinetics that is available in the United States for moderate to severe AD) and glyburide/metformin (a combination pharmacotherapy formulation approved for glycemic control in patients with type 2 diabetes mellitus). METHODS: In this single-center, multiple-dose, open-label study, healthy adult subjects received a single oral dose of memantine hydrochloride (20 mg) on day 1. After a 14-day washout period, subjects were orally administered 1.25-mg glyburide/250-mg metformin BID with food for 6 days. On day 21, subjects were coadministered memantine (20 mg) and glyburide/metformin with food. Assessments included determination of pharmacokinetic parameters for memantine and the antidiabetic agents when administered alone and in combination, pharmacodynamic measurements of blood glucose levels, and analyses of tolerability. RESULTS: The study population consisted of 24 subjects (13 women, 11 men; 79.2% white) with a mean (SD) age of 26.1 (5.6) years and a mean (SD) weight of 69.5 (11.3) kg. Twenty-one subjects completed the study: 2 discontinued due to adverse events judged unrelated to study medication, and 1 withdrew consent. No significant pharmacokinetic or pharmacodynamic interactions were observed between memantine and glyburide/metformin. Adverse events included dizziness (41.7% of patients) with memantine administration and gastrointestinal effects (nausea, 9.1 %; vomiting, 9.1%; abdominal cramps, 13.6%) with glyburide/metformin administration. CONCLUSIONS: No pharmacokinetic interactions between memantine and glyburide/metformin were detected in this study of healthy young volunteers. Memantine had no effect on the pharmacodynamic activities of glyburide and metformin, and the drug combination was well tolerated in this population.


Assuntos
Glibureto/farmacologia , Glibureto/farmacocinética , Memantina/farmacologia , Memantina/farmacocinética , Metformina/farmacologia , Metformina/farmacocinética , Adulto , Doença de Alzheimer/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Combinação de Medicamentos , Interações Medicamentosas , Feminino , Glibureto/administração & dosagem , Humanos , Masculino , Metformina/administração & dosagem
7.
Trends Neurosci ; 28(3): 152-7, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15749169

RESUMO

Although the suprachiasmatic nucleus (SCN) is well established as providing a genetically based clock for timing circadian rhythms, the mechanisms by which the timing signal is translated into circadian rhythms of behavior and underlying physiology have only recently come to light. The bulk of the SCN outflow terminates in a column of tissue that arches upward and backward from the SCN, and which includes the subparaventricular zone (SPZ) and the dorsomedial nucleus of the hypothalamus. Neurons within the dorsal SPZ are necessary for organizing circadian rhythms of body temperature, whereas neurons in the ventral SPZ are needed for circadian rhythms of sleep and waking. Ventral SPZ neurons in turn relay to the dorsomedial nucleus, which is crucial for producing circadian rhythms of sleep and waking, locomotor activity, feeding and corticosteroid production. This multistage processor provides the animal with flexibility so that environmental cues, such as food availability, ambient temperature and social interactions, can be integrated with the clock signal to sculpt an adaptive pattern of rhythmic daily activities that maximize the chances of survival and reproduction.


Assuntos
Ritmo Circadiano/fisiologia , Hipotálamo/fisiologia , Rede Nervosa/fisiologia , Animais , Meio Ambiente , Humanos , Hipotálamo/anatomia & histologia , Redes Neurais de Computação
8.
Proc Natl Acad Sci U S A ; 101(50): 17545-8, 2004 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-15583127

RESUMO

Although mammals of different species have different sleep patterns, brief sleep-wake transitions commonly are observed across species and appear to occur randomly throughout the sleeping period. The dynamical patterns and functions of these brief awakenings from sleep are not well understood, and they often are viewed as disruptions (random or pathologic) of the sleep process. In this article, we hypothesize that brief awakenings from sleep may reflect aspects of the endogenous sleep control mechanism and thus may exhibit certain robust dynamical patterns across species. We analyze sleep recordings from mice, rats, cats, and humans, and we compare the distributions of sleep and wake episode durations. For all four species, we find that durations of brief wake episodes during the sleep period exhibit a scale-free power-law behavior with an exponent alpha that remains the same for all species (alpha approximately equal to 2.2). In contrast, sleep episode durations for all four species follow exponential distributions with characteristic time scales, which change across species in relation to body mass and metabolic rate. Our findings suggest common dynamical features of brief awakenings and sleep durations across species and may provide insights into the dynamics of the neural circuits controlling sleep.


Assuntos
Mamíferos/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Peso Corporal , Encéfalo/fisiologia , Gatos , Eletrofisiologia , Humanos , Camundongos , Tamanho do Órgão , Ratos
9.
Sleep ; 27(7): 1275-81, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15586780

RESUMO

STUDY OBJECTIVES: Extensive evidence suggests that histaminergic neurons promote wakefulness. Histaminergic neurons are found exclusively in the tuberomammillary nucleus (TMN), and electrolytic lesions of the posterior hypothalamus, where the TMN resides, produce intense hypersomnolence. However, electrolytic lesions disrupt fibers of passage, and the effects of fiber-sparing, cell-specific TMN lesions on sleep and wakefulness are unknown. Hence, we placed cell-specific lesions in the TMN to determine its role in spontaneous wakefulness. DESIGN: TMN neurons in rats are relatively resistant to excitotoxins. Hence, we ablated them using saporin conjugated to hypocretin 2, which ablates hypocretin receptor-bearing neurons such as TMN neurons. One to 2 weeks after bilateral injections of Hcrt2-SAP into Sprague-Dawley rats, we correlated loss of TMN neurons with changes in sleep. SETTING: N/A. PARTICIPANTS: N/A. INTERVENTIONS: N/A. MEASUREMENTS AND RESULTS: Four days after injections with hypocretin-2-saporin, the number of TMN neurons was markedly decreased, and most were lost after 12 days, as determined by immunohistochemistry for adenosine deaminase, a marker of TMN neurons. Nearby nonhistaminergic neurons were similarly ablated. Rats with an average 82.5% loss of TMN cells (determined 2 weeks after injection) did not have marked changes in total sleep amounts compared to saline-treated rats 1 or 2 weeks following the injection, except for a slight decrease in rapid eye movement sleep during the lights-on period for the first week only. The percentage of remaining TMN neurons positively correlated with the average duration of wake bouts during the lights-off period. CONCLUSION: The absence of gross changes in sleep after extensive loss of histaminergic neurons suggests that this system is not critical for spontaneous wakefulness.


Assuntos
Histamina/fisiologia , Região Hipotalâmica Lateral/fisiologia , Sono/fisiologia , Adenosina Desaminase/metabolismo , Animais , Nível de Alerta/efeitos dos fármacos , Nível de Alerta/fisiologia , Mapeamento Encefálico , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Dominância Cerebral/efeitos dos fármacos , Dominância Cerebral/fisiologia , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/patologia , Hipotálamo Posterior/efeitos dos fármacos , Hipotálamo Posterior/patologia , Hipotálamo Posterior/fisiologia , Masculino , Vias Neurais/efeitos dos fármacos , Vias Neurais/patologia , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Vigília/efeitos dos fármacos , Vigília/fisiologia
10.
Neurosci Lett ; 370(2-3): 123-6, 2004 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-15488307

RESUMO

The lateral hypothalamic area (LHA) contains a large population of neurons that express the enzyme acetylcholinesterase (AChE), but are not themselves cholinergic. Some of these neurons have been shown to contain melanin-concentrating hormone (MCH), a neuropeptide implicated in regulating feeding, but the identities of the remaining neurons are unknown. We now report that nearly all AChE-immunoreactive neurons in the LHA express immunoreactivity for either MCH or for orexin, a peptide implicated in regulating wakefulness. Furthermore, most orexin neurons and MCH neurons appear to contain AChE. AChE immunoreactivity appears to be a key feature of nearly all of the diffusely-projecting cortical systems.


Assuntos
Acetilcolinesterase/metabolismo , Região Hipotalâmica Lateral/citologia , Hormônios Hipotalâmicos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Hormônios Hipofisários/metabolismo , Animais , Contagem de Células/métodos , Imuno-Histoquímica/métodos , Masculino , Neurônios/enzimologia , Orexinas , Ratos , Ratos Sprague-Dawley
11.
J Neurosci ; 23(33): 10691-702, 2003 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-14627654

RESUMO

The suprachiasmatic nucleus (SCN) contains the brain's circadian pacemaker, but mechanisms by which it controls circadian rhythms of sleep and related behaviors are poorly understood. Previous anatomic evidence has implicated the dorsomedial hypothalamic nucleus (DMH) in circadian control of sleep, but this hypothesis remains untested. We now show that excitotoxic lesions of the DMH reduce circadian rhythms of wakefulness, feeding, locomotor activity, and serum corticosteroid levels by 78-89% while also reducing their overall daily levels. We also show that the DMH receives both direct and indirect SCN inputs and sends a mainly GABAergic projection to the sleep-promoting ventrolateral preoptic nucleus, and a mainly glutamate-thyrotropin-releasing hormone projection to the wake-promoting lateral hypothalamic area, including orexin (hypocretin) neurons. Through these pathways, the DMH may influence a wide range of behavioral circadian rhythms.


Assuntos
Comportamento Animal/fisiologia , Ritmo Circadiano/fisiologia , Núcleo Hipotalâmico Dorsomedial/fisiologia , Corticosteroides/sangue , Animais , Comportamento Animal/efeitos dos fármacos , Toxina da Cólera/administração & dosagem , Toxina da Cólera/farmacocinética , Ritmo Circadiano/efeitos dos fármacos , Escuridão , Núcleo Hipotalâmico Dorsomedial/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/farmacocinética , Ácido Ibotênico/administração & dosagem , Masculino , Melatonina/sangue , Microinjeções , Atividade Motora/efeitos dos fármacos , Vias Neurais/fisiologia , Neurotoxinas/administração & dosagem , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Sono/fisiologia , Estilbamidinas/administração & dosagem , Estilbamidinas/farmacocinética , Núcleo Supraquiasmático/fisiologia , Vigília/efeitos dos fármacos , Vigília/fisiologia
12.
Nat Genet ; 35(2): 131-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14517540

RESUMO

We previously mapped susceptibility to stroke to chromosome 5q12. Here we finely mapped this locus and tested it for association with stroke. We found the strongest association in the gene encoding phosphodiesterase 4D (PDE4D), especially for carotid and cardiogenic stroke, the forms of stroke related to atherosclerosis. Notably, we found that haplotypes can be classified into three distinct groups: wild-type, at-risk and protective. We also observed a substantial disregulation of multiple PDE4D isoforms in affected individuals. We propose that PDE4D is involved in the pathogenesis of stroke, possibly through atherosclerosis, which is the primary pathological process underlying ischemic stroke.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/genética , Isquemia Encefálica/enzimologia , Isquemia Encefálica/genética , Cromossomos Humanos Par 5 , Desequilíbrio de Ligação , Regiões 5' não Traduzidas/genética , Sequência de Bases , Mapeamento Cromossômico , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Predisposição Genética para Doença , Humanos , Isoenzimas/genética , Repetições de Microssatélites , Dados de Sequência Molecular , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único
13.
Neuron ; 36(2): 199-211, 2002 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-12383777

RESUMO

Feeding provides substrate for energy metabolism, which is vital to the survival of every living animal and therefore is subject to intense regulation by brain homeostatic and hedonic systems. Over the last decade, our understanding of the circuits and molecules involved in this process has changed dramatically, in large part due to the availability of animal models with genetic lesions. In this review, we examine the role played in homeostatic regulation of feeding by systemic mediators such as leptin and ghrelin, which act on brain systems utilizing neuropeptide Y, agouti-related peptide, melanocortins, orexins, and melanin concentrating hormone, among other mediators. We also examine the mechanisms for taste and reward systems that provide food with its intrinsically reinforcing properties and explore the links between the homeostatic and hedonic systems that ensure intake of adequate nutrition.


Assuntos
Comportamento Alimentar/fisiologia , Homeostase/fisiologia , Filosofia , Recompensa , Animais , Encéfalo/fisiologia , Comportamento Alimentar/psicologia , Humanos
14.
Am J Hum Genet ; 71(4): 877-92, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12145742

RESUMO

The cause of schizophrenia is unknown, but it has a significant genetic component. Pharmacologic studies, studies of gene expression in man, and studies of mouse mutants suggest involvement of glutamate and dopamine neurotransmitter systems. However, so far, strong association has not been found between schizophrenia and variants of the genes encoding components of these systems. Here, we report the results of a genomewide scan of schizophrenia families in Iceland; these results support previous work, done in five populations, showing that schizophrenia maps to chromosome 8p. Extensive fine-mapping of the 8p locus and haplotype-association analysis, supplemented by a transmission/disequilibrium test, identifies neuregulin 1 (NRG1) as a candidate gene for schizophrenia. NRG1 is expressed at central nervous system synapses and has a clear role in the expression and activation of neurotransmitter receptors, including glutamate receptors. Mutant mice heterozygous for either NRG1 or its receptor, ErbB4, show a behavioral phenotype that overlaps with mouse models for schizophrenia. Furthermore, NRG1 hypomorphs have fewer functional NMDA receptors than wild-type mice. We also demonstrate that the behavioral phenotypes of the NRG1 hypomorphs are partially reversible with clozapine, an atypical antipsychotic drug used to treat schizophrenia.


Assuntos
Cromossomos Humanos Par 8 , Predisposição Genética para Doença , Neuregulina-1/genética , Esquizofrenia/genética , Animais , Mapeamento Cromossômico , Modelos Animais de Doenças , Receptores ErbB/genética , Feminino , Haplótipos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Receptor ErbB-4
15.
J Neurosci ; 22(3): 977-90, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11826126

RESUMO

Sleep is influenced by diverse factors such as circadian time, affective states, ambient temperature, pain, etc., but pathways mediating these influences are unknown. To identify pathways that may influence sleep, we examined afferents to the ventrolateral preoptic nucleus (VLPO), an area critically implicated in promoting sleep. Injections of the retrograde tracer cholera toxin B subunit (CTB) into the VLPO produced modest numbers of CTB-labeled monoaminergic neurons in the tuberomammillary nucleus, raphe nuclei, and ventrolateral medulla, as well as a few neurons in the locus coeruleus. Immunohistochemistry for monoaminergic markers showed dense innervation of the VLPO by histaminergic, noradrenergic, and serotonergic fibers. Along with previous findings, these results suggest that the VLPO and monoaminergic nuclei may be reciprocally connected. Retrograde and anterograde tracing showed moderate or heavy inputs to the VLPO from hypothalamic regions including the median preoptic nucleus, lateral hypothalamic area, and dorsomedial hypothalamic nucleus (DMH), autonomic regions including the infralimbic cortex and parabrachial nucleus, and limbic regions including the lateral septal nucleus and ventral subiculum. Light to moderate inputs arose from orexin and melanin concentrating hormone neurons, but cholinergic or dopaminergic inputs were extremely sparse. Suprachiasmatic nucleus (SCN) projections to the VLPO were sparse, but the heavy input to the VLPO from the DMH, which receives direct and indirect SCN inputs, could provide an alternate pathway regulating the circadian timing of sleep. These robust pathways suggest candidate mechanisms by which sleep may be influenced by brain systems regulating arousal, autonomic, limbic, and circadian functions.


Assuntos
Vias Aferentes/citologia , Biotina/análogos & derivados , Área Pré-Óptica/citologia , Vias Aferentes/fisiologia , Animais , Nível de Alerta/fisiologia , Transporte Axonal/fisiologia , Monoaminas Biogênicas/metabolismo , Encéfalo/citologia , Contagem de Células , Toxina da Cólera , Colina O-Acetiltransferase/metabolismo , Ritmo Circadiano/fisiologia , Dextranos , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/fisiologia , Hormônios Hipotalâmicos/metabolismo , Imuno-Histoquímica , Masculino , Melaninas/metabolismo , Neurônios/citologia , Neuropeptídeos/metabolismo , Fito-Hemaglutininas , Hormônios Hipofisários/metabolismo , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Coloração pela Prata , Sono/fisiologia , Conjugado Aglutinina do Germe de Trigo-Peroxidase do Rábano Silvestre
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...