Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Rev Neurosci ; 20(2): 83-93, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30546103

RESUMO

Narcolepsy is the most common neurological cause of chronic sleepiness. The discovery about 20 years ago that narcolepsy is caused by selective loss of the neurons producing orexins (also known as hypocretins) sparked great advances in the field. Here, we review the current understanding of how orexin neurons regulate sleep-wake behaviour and the consequences of the loss of orexin neurons. We also summarize the developing evidence that narcolepsy is an autoimmune disorder that may be caused by a T cell-mediated attack on the orexin neurons and explain how these new perspectives can inform better therapeutic approaches.


Assuntos
Narcolepsia/fisiopatologia , Sistema Nervoso/fisiopatologia , Animais , Doenças Autoimunes , Humanos , Narcolepsia/metabolismo , Narcolepsia/patologia , Sistema Nervoso/metabolismo , Sistema Nervoso/patologia , Neurobiologia , Orexinas/metabolismo
2.
J Neurosci ; 37(15): 3995-4006, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28235898

RESUMO

Narcolepsy is characterized by chronic sleepiness and cataplexy-sudden muscle paralysis triggered by strong, positive emotions. This condition is caused by a lack of orexin (hypocretin) signaling, but little is known about the neural mechanisms that mediate cataplexy. The amygdala regulates responses to rewarding stimuli and contains neurons active during cataplexy. In addition, lesions of the amygdala reduce cataplexy. Because GABAergic neurons of the central nucleus of the amygdala (CeA) target brainstem regions known to regulate muscle tone, we hypothesized that these cells promote emotion-triggered cataplexy. We injected adeno-associated viral vectors coding for Cre-dependent DREADDs or a control vector into the CeA of orexin knock-out mice crossed with vGAT-Cre mice, resulting in selective expression of the excitatory hM3 receptor or the inhibitory hM4 receptor in GABAergic neurons of the CeA. We measured sleep/wake behavior and cataplexy after injection of saline or the hM3/hM4 ligand clozapine-N-oxide (CNO) under baseline conditions and under conditions that should elicit positive emotions. In mice expressing hM3, CNO approximately doubled the amount of cataplexy in the first 3 h after dosing under baseline conditions. Rewarding stimuli (chocolate or running wheels) also increased cataplexy, but CNO produced no further increase. In mice expressing hM4, CNO reduced cataplexy in the presence of chocolate or running wheels. These results demonstrate that GABAergic neurons of the CeA are sufficient and necessary for the production of cataplexy in mice, and they likely are a key part of the mechanism through which positive emotions trigger cataplexy.SIGNIFICANCE STATEMENT Cataplexy is one of the major symptoms of narcolepsy, but little is known about how strong, positive emotions trigger these episodes of muscle paralysis. Prior research shows that amygdala neurons are active during cataplexy and cataplexy is reduced by lesions of the amygdala. We found that cataplexy is substantially increased by selective activation of GABAergic neurons in the central nucleus of the amygdala (CeA). We also demonstrate that inhibition of these neurons reduces reward-promoted cataplexy. These results build upon prior work to establish the CeA as a crucial element in the neural mechanisms of cataplexy. These results demonstrate the importance of the CeA in regulating responses to rewarding stimuli, shedding light on the broader neurobiology of emotions and motor control.


Assuntos
Cataplexia/genética , Cataplexia/metabolismo , Núcleo Central da Amígdala/metabolismo , Neurônios GABAérgicos/metabolismo , Animais , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Knockout
3.
J Neurosci ; 37(5): 1352-1366, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28039375

RESUMO

The pedunculopontine tegmental (PPT) nucleus has long been implicated in the regulation of cortical activity and behavioral states, including rapid eye-movement (REM) sleep. For example, electrical stimulation of the PPT region during sleep leads to rapid awakening, whereas lesions of the PPT in cats reduce REM sleep. Though these effects have been linked with the activity of cholinergic PPT neurons, the PPT also includes intermingled glutamatergic and GABAergic cell populations, and the precise roles of cholinergic, glutamatergic, and GABAergic PPT cell groups in regulating cortical activity and behavioral state remain unknown. Using a chemogenetic approach in three Cre-driver mouse lines, we found that selective activation of glutamatergic PPT neurons induced prolonged cortical activation and behavioral wakefulness, whereas inhibition reduced wakefulness and increased non-REM (NREM) sleep. Activation of cholinergic PPT neurons suppressed lower-frequency electroencephalogram rhythms during NREM sleep. Last, activation of GABAergic PPT neurons slightly reduced REM sleep. These findings reveal that glutamatergic, cholinergic, and GABAergic PPT neurons differentially influence cortical activity and sleep/wake states. SIGNIFICANCE STATEMENT: More than 40 million Americans suffer from chronic sleep disruption, and the development of effective treatments requires a more detailed understanding of the neuronal mechanisms controlling sleep and arousal. The pedunculopontine tegmental (PPT) nucleus has long been considered a key site for regulating wakefulness and REM sleep. This is mainly because of the cholinergic neurons contained in the PPT nucleus. However, the PPT nucleus also contains glutamatergic and GABAergic neurons that likely contribute to the regulation of cortical activity and sleep-wake states. The chemogenetic experiments in the present study reveal that cholinergic, glutamatergic, and GABAergic PPT neurons each have distinct effects on sleep/wake behavior, improving our understanding of how the PPT nucleus regulates cortical activity and behavioral states.


Assuntos
Neurônios Colinérgicos/fisiologia , Neurônios GABAérgicos/fisiologia , Glutamatos/fisiologia , Neurônios/fisiologia , Núcleo Tegmental Pedunculopontino/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Animal/fisiologia , Eletroencefalografia , Eletromiografia , Camundongos , Núcleo Tegmental Pedunculopontino/citologia , Sono REM/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
4.
Neurobiol Dis ; 120: 12-20, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30149182

RESUMO

The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Narcolepsia/metabolismo , Neurônios/fisiologia , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Feminino , Hormônios Hipotalâmicos/antagonistas & inibidores , Hormônios Hipotalâmicos/genética , Masculino , Melaninas/antagonistas & inibidores , Melaninas/genética , Camundongos , Camundongos Knockout , Narcolepsia/genética , Neurônios/efeitos dos fármacos , Piperidinas/farmacologia , Hormônios Hipofisários/antagonistas & inibidores , Hormônios Hipofisários/genética
5.
Am J Physiol Regul Integr Comp Physiol ; 310(1): R41-54, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491097

RESUMO

The parabrachial nucleus is important for thermoregulation because it relays skin temperature information from the spinal cord to the hypothalamus. Prior work in rats localized thermosensory relay neurons to its lateral subdivision (LPB), but the genetic and neurochemical identity of these neurons remains unknown. To determine the identity of LPB thermosensory neurons, we exposed mice to a warm (36°C) or cool (4°C) ambient temperature. Each condition activated neurons in distinct LPB subregions that receive input from the spinal cord. Most c-Fos+ neurons in these LPB subregions expressed the transcription factor marker FoxP2. Consistent with prior evidence that LPB thermosensory relay neurons are glutamatergic, all FoxP2+ neurons in these subregions colocalized with green fluorescent protein (GFP) in reporter mice for Vglut2, but not for Vgat. Prodynorphin (Pdyn)-expressing neurons were identified using a GFP reporter mouse and formed a caudal subset of LPB FoxP2+ neurons, primarily in the dorsal lateral subnucleus (PBdL). Warm exposure activated many FoxP2+ neurons within PBdL. Half of the c-Fos+ neurons in PBdL were Pdyn+, and most of these project into the preoptic area. Cool exposure activated a separate FoxP2+ cluster of neurons in the far-rostral LPB, which we named the rostral-to-external lateral subnucleus (PBreL). These findings improve our understanding of LPB organization and reveal that Pdyn-IRES-Cre mice provide genetic access to warm-activated, FoxP2+ glutamatergic neurons in PBdL, many of which project to the hypothalamus.


Assuntos
Febre/metabolismo , Hipotermia/metabolismo , Neurônios/metabolismo , Núcleos Parabraquiais/metabolismo , Temperatura Cutânea , Sensação Térmica , Animais , Modelos Animais de Doenças , Encefalinas/genética , Encefalinas/metabolismo , Febre/genética , Febre/fisiopatologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Genótipo , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipotermia/genética , Hipotermia/fisiopatologia , Integrases/genética , Integrases/metabolismo , Sítios Internos de Entrada Ribossomal , Masculino , Camundongos Transgênicos , Técnicas de Rastreamento Neuroanatômico , Núcleos Parabraquiais/fisiopatologia , Fenótipo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Repressoras/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
6.
Front Behav Neurosci ; 16: 837523, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35401134

RESUMO

Normal neurodevelopment requires precise expression of the key ubiquitin ligase gene Ube3a. Comparing newly generated mouse models for Ube3a downregulation (models of Angelman syndrome) vs. Ube3a upregulation (models for autism), we find reciprocal effects of Ube3a gene dosage on phenotypes associated with circadian rhythmicity, including the amount of locomotor activity. Consistent with results from neurons in general, we find that Ube3a is imprinted in neurons of the suprachiasmatic nuclei (SCN), the pacemaking circadian brain locus, despite other claims that SCN neurons were somehow exceptional to these imprinting rules. In addition, Ube3a-deficient mice lack the typical drop in wake late in the dark period and have blunted responses to sleep deprivation. Suppression of physical activity by light in Ube3a-deficient mice is not due to anxiety as measured by behavioral tests and stress hormones; quantification of stress hormones may provide a mechanistic link to sleep alteration and memory deficits caused by Ube3a deficiency, and serve as an easily measurable biomarker for evaluating potential therapeutic treatments for Angelman syndrome. We conclude that reduced Ube3a gene dosage affects not only neurodevelopment but also sleep patterns and circadian rhythms.

7.
Am J Physiol Regul Integr Comp Physiol ; 299(3): R751-61, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20592176

RESUMO

To evaluate the contribution of neural pathways to the determination of the circadian oscillator phase in peripheral organs, we assessed lateralization of clock gene expression in Syrian hamsters induced to split rhythms of locomotor activity by exposure to constant light. We measured the ratio of haPer1, haPer2, and haBmal1 mRNA on the high vs. low (H/L) side at 3-h intervals prior to the predicted activity onset (pAO). We also calculated expression on the sides ipsilateral vs. contralateral (I/C) to the side of the suprachiasmatic nucleus (SCN) expressing higher haPer1. The extent of asymmetry in split hamsters varied between specific genes, phases, and organs. Although the magnitude of asymmetry in peripheral organs was never as great as that in the SCN, we observed significantly greater lateralization of clock gene expression in the adrenal medulla and cortex, lung, and skeletal muscle, but not in liver or kidney, of split hamsters than of unsplit controls. We observed fivefold lateralization of expression of the clock-controlled gene, albumin site D-element binding protein (Dbp), in skeletal muscle (H/L: 10.7 +/- 3.7 at 3 h vs. 2.2 +/- 0.3 at 0 h pAO; P = 0.03). Furthermore, tyrosine hydroxylase expression was asymmetrical in the adrenal medulla of split (H/L: 1.9 +/- 0.5 at 0 h) vs. unsplit hamsters (1.2 +/- 0.04; P < 0.05). Consistent with a model of neurally controlled gene expression, we found significant correlations between the phase angle between morning and evening components (psi(me)) and the level of asymmetry (H/L or I/C). Our results indicate that neural pathways contribute to, but cannot completely account for, SCN regulation of the phase of peripheral oscillators.


Assuntos
Ritmo Circadiano/fisiologia , Vias Neurais/fisiologia , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/fisiologia , Animais , Cricetinae , Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Mesocricetus , Atividade Motora/fisiologia , Músculo Esquelético/metabolismo , Especificidade de Órgãos , Proteínas Circadianas Period/genética , Fotoperíodo
8.
Sleep ; 43(6)2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31830270

RESUMO

Orexin receptor antagonists are clinically useful for treating insomnia, but thorough blockade of orexin signaling could cause narcolepsy-like symptoms. Specifically, while sleepiness is a desirable effect, an orexin antagonist could also produce cataplexy, sudden episodes of muscle weakness often triggered by strong, positive emotions. In this study, we examined the effects of dual orexin receptor antagonists (DORAs), lemborexant (E2006) and almorexant, on sleep-wake behavior and cataplexy during the dark period in wild-type (WT) mice and prepro-orexin knockout (OXKO) mice. In WT mice, lemborexant at 10 and 30 mg/kg quickly induced NREM sleep in a dose-dependent fashion. In contrast, lemborexant did not alter sleep-wake behavior in OXKO mice. Under the baseline condition, cataplexy was rare in lemborexant-treated WT mice, but when mice were given chocolate as a rewarding stimulus, lemborexant dose-dependently increased cataplexy. Almorexant produced similar results. Collectively, these results demonstrate that DORAs potently increase NREM and REM sleep in mice via blockade of orexin signaling, and higher doses can cause cataplexy when co-administered with a likely rewarding stimulus.


Assuntos
Cataplexia , Distúrbios do Início e da Manutenção do Sono , Animais , Cataplexia/tratamento farmacológico , Camundongos , Camundongos Knockout , Antagonistas dos Receptores de Orexina/farmacologia , Antagonistas dos Receptores de Orexina/uso terapêutico , Receptores de Orexina , Orexinas/farmacologia , Sono , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Vigília
10.
J Comp Neurol ; 525(7): 1668-1684, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-27997037

RESUMO

The orexin (hypocretin) neurons play an essential role in promoting arousal, and loss of the orexin neurons results in narcolepsy, a condition characterized by chronic sleepiness and cataplexy. The orexin neurons excite wake-promoting neurons in the basal forebrain (BF), and a reciprocal projection from the BF back to the orexin neurons may help promote arousal and motivation. The BF contains at least three different cell types (cholinergic, glutamatergic, and γ-aminobutyric acid (GABA)ergic neurons) across its different regions (medial septum, diagonal band, magnocellular preoptic area, and substantia innominata). Given the neurochemical and anatomical heterogeneity of the BF, we mapped the pattern of BF projections to the orexin neurons across multiple BF regions and neuronal types. We performed conditional anterograde tracing using mice that express Cre recombinase only in neurons producing acetylcholine, glutamate, or GABA. We found that the orexin neurons are heavily apposed by axon terminals of glutamatergic and GABAergic neurons of the substantia innominata (SI) and magnocellular preoptic area, but there was no innervation by the cholinergic neurons. Channelrhodopsin-assisted circuit mapping (CRACM) demonstrated that glutamatergic SI neurons frequently form functional synapses with the orexin neurons, but, surprisingly, functional synapses from SI GABAergic neurons were rare. Considering their strong reciprocal connections, BF and orexin neurons likely work in concert to promote arousal, motivation, and other behaviors. J. Comp. Neurol. 525:1668-1684, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Prosencéfalo Basal/citologia , Vias Neurais/citologia , Neurônios/citologia , Animais , Prosencéfalo Basal/metabolismo , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Microscopia Confocal , Vias Neurais/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo
11.
Med Sci Sports Exerc ; 46(1): 156-66, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23846160

RESUMO

INTRODUCTION: Competitive female athletes restrict energy intake and increase exercise energy expenditure frequently resulting in ovarian suppression. The purpose of this study was to determine the impact of ovarian suppression and energy deficit on swimming performance (400-m swim velocity). METHODS: Menstrual status was determined by circulating estradiol (E2) and progesterone (P4) in ten junior elite female swimmers (15-17 yr). The athletes were categorized as cyclic (CYC) or ovarian-suppressed (OVS). They were evaluated every 2 wk for metabolic hormones, bioenergetic parameters, and sport performance during the 12-wk season. RESULTS: CYC and OVS athletes were similar (P > 0.05) in age (CYC = 16.2 ± 1.8 yr, OVS = 17 ± 1.7 yr), body mass index (CYC = 21 ± 0.4 kg·m, OVS = 25 ± 0.8 kg·m), and gynecological age (CYC = 2.6 ± 1.1 yr, OVS = 2.8 ± 1.5 yr). OVS had suppressed P4 (P < 0.001) and E2 (P = 0.002) across the season. Total triiodothyronine (TT3) and insulin-like growth factor (IGF-1) were lower in OVS (TT3: CYC = 1.6 ± 0.2 nmol·L, OVS = 1.4 ± 0.1 nmol·L, P < 0.001; IGF-1: CYC = 243 ± 1 µg·mL, OVS = 214 ± 3 µg·mL P < 0.001) than CYC at week 12. Energy intake (P < 0.001) and energy availability (P < 0.001) were significantly lower in OVS versus CYC. OVS exhibited a 9.8% decline in Δ400-m swim velocity compared with an 8.2% improvement in CYC at week 12. CONCLUSIONS: Ovarian steroids (P4 and E2), metabolic hormones (TT3 and IGF-1), and energy status markers (EA and EI) were highly correlated with sport performance. This study illustrates that when exercise training occurs in the presence of ovarian suppression with evidence for energy conservation (i.e., reduced TT3), it is associated with poor sport performance. These data from junior elite female athletes support the need for dietary periodization to help optimize energy intake for appropriate training adaptation and maximal sport performance.


Assuntos
Desempenho Atlético/fisiologia , Insuficiência Ovariana Primária/fisiopatologia , Natação/fisiologia , Adolescente , Ingestão de Energia/fisiologia , Metabolismo Energético/fisiologia , Estradiol/sangue , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Ciclo Menstrual/sangue , Ciclo Menstrual/fisiologia , Insuficiência Ovariana Primária/sangue , Progesterona/sangue , Tri-Iodotironina/sangue
12.
PLoS One ; 8(6): e67173, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826226

RESUMO

Cells of the dorsomedial/lateral hypothalamus (DMH/LH) that produce hypocretin (HCRT) promote arousal in part by activation of cells of the locus coeruleus (LC) which express tyrosine hydroxylase (TH). The suprachiasmatic nucleus (SCN) drives endogenous daily rhythms, including those of sleep and wakefulness. These circadian oscillations are generated by a transcriptional-translational feedback loop in which the Period (Per) genes constitute critical components. This cell-autonomous molecular clock operates not only within the SCN but also in neurons of other brain regions. However, the phenotype of such neurons and the nature of the phase controlling signal from the pacemaker are largely unknown. We used dual fluorescent in situ hybridization to assess clock function in vasopressin, HCRT and TH cells of the SCN, DMH/LH and LC, respectively, of male Syrian hamsters. In the first experiment, we found that Per1 expression in HCRT and TH oscillated in animals held in constant darkness with a peak phase that lagged that in AVP cells of the SCN by several hours. In the second experiment, hamsters induced to split their locomotor rhythms by exposure to constant light had asymmetric Per1 expression within cells of the middle SCN at 6 h before activity onset (AO) and in HCRT cells 9 h before and at AO. We did not observe evidence of lateralization of Per1 expression in the LC. We conclude that the SCN communicates circadian phase to HCRT cells via lateralized neural projections, and suggests that Per1 expression in the LC may be regulated by signals of a global or bilateral nature.


Assuntos
Nível de Alerta/fisiologia , Ritmo Circadiano/fisiologia , Neurônios/fisiologia , Animais , Cricetinae , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Locus Cerúleo/metabolismo , Masculino , Mesocricetus , Neuropeptídeos/metabolismo , Orexinas , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
13.
Endocrinology ; 150(3): 1202-16, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19008313

RESUMO

Orexin-A (ORXA) is an orexigenic neuropeptide produced by the lateral hypothalamus that increases food intake when injected into the brain ventricles or forebrain nuclei. We used a licking microstructure analysis to evaluate hindbrain and forebrain ORXA effects in intact and hindbrain-lesioned rats, to identify the motivational and anatomical bases of ORXA hyperphagia. Intact rats with cannulas in the fourth brain ventricle (4V) received vehicle (artificial cerebrospinal fluid) or ORXA (0.1, 0.4, 1, or 10 nm) injections before 90 min access to 0.1 m sucrose. Meal size and frequency were increased in a double-dissociated manner by the 1 and 10 nm doses, respectively. In experiment 2, 4V 1 nm ORXA was applied to rats offered solutions varied in caloric and gustatory intensity (water and 0.1 and 1 m sucrose). ORXA increased meal frequency for all tastants. ORXA increased meal size only for 0.1 m sucrose, by prolonging the meal without affecting early ingestion rate or lick burst size, suggesting that 4V ORXA influenced inhibitory postingestive feedback rather than taste evaluation. In experiment 3, rats with cannulas in the third ventricle (3V) received dorsal medullary lesions centered on the area postrema (APX group) or sham procedures, and licking for water and 0.1 and 1 m sucrose was evaluated after 1 nm 3V ORXA/artificial cerebrospinal fluid injections. The 3V ORXA increased 0.1 m sucrose meal size and meal frequency for all tastants in the sham group, as observed after 4V ORXA in experiment 2. In the APX group, 3V ORXA injections influenced meal frequency, but they no longer increased meal size. However, the APX rats increased meal size for 0.1 m sucrose after food and water deprivation and after 3V angiotensin II injection. They also showed meal size suppression after 3V injection of the melanocortin-3/4 receptor agonist melanotan II (1 nm). These findings suggest that the area postrema and subjacent nucleus of the solitary tract are necessary for increases in consummatory (meal size) but not appetitive (meal frequency) responses to 3V ORXA. The meal size increases may be due to reduced postingestive feedback inhibition induced by ORXA delivered to either the hindbrain or forebrain ventricles.


Assuntos
Comportamento Alimentar/efeitos dos fármacos , Hiperfagia/induzido quimicamente , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neuropeptídeos/farmacologia , Rombencéfalo/efeitos dos fármacos , Animais , Apetite/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Bombas de Infusão , Injeções Intraventriculares , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Masculino , Neuropeptídeos/administração & dosagem , Orexinas , Ratos , Ratos Sprague-Dawley , Rombencéfalo/fisiologia , Paladar/efeitos dos fármacos
14.
Am J Physiol Regul Integr Comp Physiol ; 294(2): R329-43, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17989139

RESUMO

Melanin-concentrating hormone (MCH) and neuropeptide Y (NPY) are orexigenic peptides found in hypothalamic neurons that project throughout the forebrain and hindbrain. The effects of fourth ventricle (4V) infusions of NPY (5 microg) and MCH (5 microg) on licking for water, 4 mM saccharin, and sucrose (0.1 and 1.0 M) solutions were compared to identify the contributions of each peptide to hindbrain-stimulated feeding. NPY increased mean meal size only for the sucrose solutions, suggesting that caloric feedback or taste quality is pertinent to the orexigenic effect; MCH infusions under identical testing conditions failed to produce increases for any tastant. A second experiment also observed no intake or licking effects after MCH doses up to 15 microg, supporting the conclusion that MCH-induced orexigenic responses require forebrain stimulation. A third experiment compared the 4V NPY results with those obtained after NPY infusions (5 microg) into the third ventricle (3V). In contrast to the effects observed after the 3V NPY injections and previously reported forebrain intracerebroventricular (ICV) NPY infusion studies, 4V NPY failed to increase meal frequency for any taste solution or ingestion rate in the early phases of the sucrose meals. Overall, 4V NPY responses were limited to intrameal behavioral processes, whereas forebrain ICV NPY stimulation elicited both consummatory and appetitive responses. The dissociation between MCH and NPY effects observed for 4V injections is consistent with reports that forebrain ICV injections of MCH and NPY produced nearly dichotomous effects on the pattern of licking microstructure, and, collectively, the results indicate that the two peptides have separate sites of feeding action in the brain.


Assuntos
Comportamento de Ingestão de Líquido/fisiologia , Comportamento Alimentar/fisiologia , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Neuropeptídeo Y/metabolismo , Hormônios Hipofisários/metabolismo , Rombencéfalo/fisiologia , Animais , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Líquidos/fisiologia , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Hormônios Hipotalâmicos/farmacologia , Masculino , Melaninas/farmacologia , Neuropeptídeo Y/farmacologia , Hormônios Hipofisários/farmacologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/fisiologia , Ratos , Ratos Sprague-Dawley , Rombencéfalo/efeitos dos fármacos , Sacarina/farmacologia , Sacarose/farmacologia , Paladar , Água/farmacologia
15.
Curr Sports Med Rep ; 6(3): 190-4, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19202666

RESUMO

Competitive female athletes face many challenges unlike their recreationally active counterparts. As sport has advanced for girls and women, the physical, psychologic, and nutritional demands force female athletes to develop optimal strategies for competitive success. Sports medicine professionals must consume volumes of research investigating issues regarding competitive female athletes. This review focuses on three primary factors associated with females and performance: iron status, interrelated biorhythms, and energy optimization. Consideration of these factors in both health and performance goals is critical to the long-term success of competitive female athletes.


Assuntos
Desempenho Atlético/fisiologia , Esportes/fisiologia , Anemia Ferropriva/fisiopatologia , Feminino , Ferritinas/sangue , Humanos , Ciclo Menstrual/fisiologia , Distúrbios Menstruais/fisiopatologia , Distúrbios Menstruais/terapia
16.
J Strength Cond Res ; 18(2): 302-5, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15142018

RESUMO

Open-water swimming (5, 10, and 25 km) has many unique challenges that separate it from other endurance sports, like marathon running and cycling. The characteristics of a successful open-water swimmer are unclear. The purpose of this study was to determine the physical and metabolic characteristics of a group of elite-level open-water swimmers. The open-water swimmers were participating in a 1-week training camp. Anthropometric, metabolic, and blood chemistry assessments were performed on the athletes. The swimmers had a VO(2)peak of 5.51 +/- 0.96 and 5.06 +/- 0.57 ml.kg(-1).min(-1) for males and females, respectively. Their lactate threshold (LT) occurred at a pace equal to 88.75% of peak pace for males and 93.75% for females. These elite open-water swimmers were smaller and lighter than competitive pool swimmers. They possess aerobic metabolic alterations that resulted in enhanced performance in distance swimming. Trainers and coaches should develop dry-land programs that will improve the athlete's muscular endurance. Furthermore, programs should be designed to increase the LT velocity as a percentage of peak swimming velocity.


Assuntos
Antropometria , Metabolismo , Natação/fisiologia , Adolescente , Limiar Anaeróbio , Análise Química do Sangue , Feminino , Humanos , Masculino , Consumo de Oxigênio , Valores de Referência , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA