Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Eur J Histochem ; 65(s1)2021 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-34755506

RESUMO

Bisphenol A (BPA), an organic synthetic compound found in some plastics and epoxy resins, is classified as an endocrine disrupting chemical. Exposure to BPA is especially dangerous if it occurs during specific "critical periods" of life, when organisms are more sensitive to hormonal changes (i.e., intrauterine, perinatal, juvenile or puberty periods). In this study, we focused on the effects of chronic exposure to BPA in adult female mice starting during pregnancy. Three months old C57BL/6J females were orally exposed to BPA or to vehicle (corn oil). The treatment (4 µg/kg body weight/day) started the day 0 of pregnancy and continued throughout pregnancy, lactation, and lasted for a total of 20 weeks. BPA-treated dams did not show differences in body weight or food intake, but they showed an altered estrous cycle compared to the controls. In order to evidence alterations in social and sociosexual behaviors, we performed the Three-Chamber test for sociability, and analyzed two hypothalamic circuits (well-known targets of endocrine disruption) particularly involved in the control of social behavior: the vasopressin and the oxytocin systems. The test revealed some alterations in the displaying of social behavior: BPA-treated dams have higher locomotor activity compared to the control dams, probably a signal of high level of anxiety. In addition, BPA-treated dams spent more time interacting with no-tester females than with no-tester males. In brain sections, we observed a decrease of vasopressin immunoreactivity (only in the paraventricular and suprachiasmatic nuclei) of BPA-treated females, while we did not find any alteration of the oxytocin system. In parallel, we have also observed, in the same hypothalamic nuclei, a significant reduction of the membrane estrogen receptor GPER1 expression.


Assuntos
Comportamento Animal/efeitos dos fármacos , Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Fenóis/toxicidade , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Vasopressinas/metabolismo , Animais , Ciclo Estral/efeitos dos fármacos , Feminino , Masculino , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/patologia , Gravidez , Comportamento Social , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/patologia
2.
World Neurosurg ; 155: e460-e471, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34454071

RESUMO

BACKGROUND: Endoscopic endonasal surgery has proved to offer a practical route to treat suprasellar lesions, including tumors and vascular pathologies. Understanding the different configurations of the anterior cerebral communicating artery (ACoA) complex (ACoA-C) is crucial to properly navigate the suprachiasmatic space and decrease any vascular injury while approaching this region through an endonasal approach. METHODS: An endoscopic endonasal transplanum-transtubercular approach was performed on 36 cadaveric heads (72 sides). The variations of the ACoA-C and feasibility of reaching its different components were analyzed. The surgical area exposure of the lamina terminalis was also quantified before and after mobilization of the ACoA-C. RESULTS: The typical ACoA-C configuration was found in 41.6% of specimens. The following 2 main variations were identified: accessory anterior cerebral artery segment 2 (5, 13.9%) and common trunk of anterior cerebral artery with absence of ACoA (5, 13.9%). Of 101 recurrent arteries of Heubner, 96 (95.0%) were identified within 4 mm proximal or distal to the ACoA. The mean lamina terminalis exposure area was 33.1 ± 16.7 mm2, which increased to 59.9 ± 11.9 mm2 after elevating the ACoA. CONCLUSIONS: A considerable amount of variation of the ACoA-C can be found through an endoscopic endonasal transplanum-transtubercular approach. These configurations determine the feasibility of lamina terminalis exposure and the complexity of reaching the ACoA. Assessment of ACoA morphology and its adjacent structures is crucial while approaching the suprachiasmatic through a transnasal corridor.


Assuntos
Artérias Cerebrais/cirurgia , Cavidade Nasal/cirurgia , Neuroendoscopia/métodos , Núcleo Supraquiasmático/irrigação sanguínea , Núcleo Supraquiasmático/cirurgia , Cadáver , Artérias Cerebrais/patologia , Humanos , Cavidade Nasal/patologia , Núcleo Supraquiasmático/patologia
3.
Neuroscience ; 413: 264-278, 2019 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-31254543

RESUMO

Repetitive mild traumatic brain injury (RmTBI) is a prevalent and costly head injury particularly among adolescents. These injuries may result in long-term consequences, especially during this critical period of development. Insomnia and sleeping difficulties are frequently reported following RmTBI and greatly impair recovery. We sought to develop an animal model of exacerbated deficits following RmTBI by disrupting the hypothalamic circadian system. To accomplish this, we conducted RmTBI on adolescent rats that had received neonatal injections of monosodium glutamate (MSG), a known hypothalamic neurotoxin. We then examined behavioral, circadian, and epigenetic changes. MSG treated rats showed lower anxiety-like behaviors and displayed poor short-term working memory. We also showed changes in the morphology of the circadian clock in the suprachiasmatic nucleus (SCN) vasoactive intestinal polypeptide (VIP) immunostaining. VIP optical density in the SCN increased with MSG but decreased with RmTBI. There were changes in the expression of the clock genes and upregulation of the orexin receptors in response to RmTBI. MSG treated rats had longer telomere lengths than controls. Finally, although both MSG and RmTBI alone produced attenuated circadian amplitudes of activity and body temperature, exacerbated deficits were not identified in animals that received MSG and RmTBI. In sum, both MSG and RmTBI can alter behavior, circadian rhythm amplitude, SCN morphology, and gene expression independently, but the effects do not appear to be additive. Specific damage in the hypothalamus and SCN should be considered when patients experience sleeping problems following RmTBI, as this may improve therapeutic strategies.


Assuntos
Concussão Encefálica/metabolismo , Hipotálamo/metabolismo , Animais , Ansiedade/metabolismo , Ansiedade/patologia , Temperatura Corporal , Concussão Encefálica/patologia , Ritmo Circadiano/fisiologia , Feminino , Expressão Gênica , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/patologia , Masculino , Memória de Curto Prazo/fisiologia , Atividade Motora/fisiologia , Distribuição Aleatória , Ratos Sprague-Dawley , Recidiva , Glutamato de Sódio/efeitos adversos , Núcleo Supraquiasmático/crescimento & desenvolvimento , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia , Telômero
4.
Am J Physiol Endocrinol Metab ; 313(2): E213-E221, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28465284

RESUMO

Metabolic state and circadian clock function exhibit a complex bidirectional relationship. Circadian disruption increases propensity for metabolic dysfunction, whereas common metabolic disorders such as obesity and type 2 diabetes (T2DM) are associated with impaired circadian rhythms. Specifically, alterations in glucose availability and glucose metabolism have been shown to modulate clock gene expression and function in vitro; however, to date, it is unknown whether development of diabetes imparts deleterious effects on the suprachiasmatic nucleus (SCN) circadian clock and SCN-driven outputs in vivo. To address this question, we undertook studies in aged diabetic rats transgenic for human islet amyloid polypeptide, an established nonobese model of T2DM (HIP rat), which develops metabolic defects closely recapitulating those present in patients with T2DM. HIP rats were also cross-bred with a clock gene reporter rat model (Per1:luciferase transgenic rat) to permit assessment of the SCN and the peripheral molecular clock function ex vivo. Utilizing these animal models, we examined effects of diabetes on 1) behavioral circadian rhythms, 2) photic entrainment of circadian activity, 3) SCN and peripheral tissue molecular clock function, and 4) melatonin secretion. We report that circadian activity, light-induced entrainment, molecular clockwork, as well as melatonin secretion are preserved in the HIP rat model of T2DM. These results suggest that despite the well-characterized ability of glucose to modulate circadian clock gene expression acutely in vitro, SCN clock function and key behavioral and physiological outputs appear to be preserved under chronic diabetic conditions characteristic of nonobese T2DM.


Assuntos
Comportamento Animal/fisiologia , Ritmo Circadiano/genética , Diabetes Mellitus Tipo 2 , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Luz , Masculino , Proteínas Circadianas Period/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia
5.
Exp Neurol ; 294: 58-67, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28461004

RESUMO

Disruption of normal circadian rhythm physiology is associated with neurodegenerative disease, which can lead to symptoms such as altered sleep cycles. In Alzheimer's disease (AD), circadian dysfunction has been attributed to ß-amyloidosis. However, it is unclear whether tauopathy, another AD-associated neuropathology, can disrupt the circadian clock. We have evaluated the status of the circadian clock in a mouse model of tauopathy (Tg4510). Tg4510 mice display a long free-running period at an age when tauopathy is present, and show evidence of tauopathy in the suprachiasmatic nucleus (SCN) of the hypothalamus - the site of the master circadian clock. Additionally, cyclic expression of the core clock protein PER2 is disrupted in the hypothalamus of Tg4510 mice. Finally, disruption of the cyclic expression of PER2 and BMAL1, another core circadian clock protein, is evident in the Tg4510 hippocampus. These results demonstrate that tauopathy disrupts normal circadian clock function both at the behavioral and molecular levels, which may be attributed to the tauopathy-induced neuropathology in the SCN. Furthermore, these results establish the Tg4510 mouse line as a model to study how tauopathy disrupts normal circadian rhythm biology.


Assuntos
Transtornos Cronobiológicos/etiologia , Tauopatias/complicações , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Análise de Variância , Animais , Transtornos Cronobiológicos/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Locomoção/genética , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fosforilação/genética , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética , Proteínas tau/metabolismo
6.
eNeuro ; 4(2)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28374011

RESUMO

The suprachiasmatic nucleus (SCN) is generally considered the master clock, independently driving all circadian rhythms. We recently demonstrated the SCN receives metabolic and cardiovascular feedback adeptly altering its neuronal activity. In the present study, we show that microcuts effectively removing SCN-arcuate nucleus (ARC) interconnectivity in Wistar rats result in a loss of rhythmicity in locomotor activity, corticosterone levels, and body temperature in constant dark (DD) conditions. Elimination of these reciprocal connections did not affect SCN clock gene rhythmicity but did cause the ARC to desynchronize. Moreover, unilateral SCN lesions with contralateral retrochiasmatic microcuts resulted in identical arrhythmicity, proving that for the expression of physiological rhythms this reciprocal SCN-ARC interaction is essential. The unaltered SCN c-Fos expression following glucose administration in disconnected animals as compared to a significant decrease in controls demonstrates the importance of the ARC as metabolic modulator of SCN neuronal activity. Together, these results indicate that the SCN is more than an autonomous clock, and forms an essential component of a larger network controlling homeostasis. The present novel findings illustrate how an imbalance between SCN and ARC communication through circadian disruption could be involved in the etiology of metabolic disorders.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Ritmo Circadiano/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Temperatura Corporal/fisiologia , Corticosterona/metabolismo , Glucose/administração & dosagem , Glucose/metabolismo , Fígado/metabolismo , Modelos Animais , Atividade Motora/fisiologia , Vias Neurais/fisiologia , Vias Neurais/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas Circadianas Period/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar , Núcleo Supraquiasmático/patologia , Núcleo Supraquiasmático/fisiopatologia
7.
Eur Arch Psychiatry Clin Neurosci ; 267(5): 427-443, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28035472

RESUMO

The vasopressin- and oxytocin-degrading enzyme insulin-regulated aminopeptidase (IRAP) is expressed in various organs including the brain. However, knowledge about its presence in human hypothalamus is fragmentary. Functionally, for a number of reasons (genetic linkage, hydrolysis of oxytocin and vasopressin, its role as angiotensin IV receptor in learning and memory and others) IRAP might play a role in schizophrenia. We studied the regional and cellular localization of IRAP in normal human brain with special emphasis on the hypothalamus and determined numerical densities of IRAP-expressing cells in the paraventricular, supraoptic and suprachiasmatic nuclei in schizophrenia patients and controls. By using immunohistochemistry and Western blot analysis, IRAP was immunolocalized in postmortem human brains. Cell countings were performed to estimate numbers and numerical densities of IRAP immunoreactive hypothalamic neurons in schizophrenia patients and control cases. Shape, size and regional distribution of IRAP-expressing cells, as well the lack of co-localization with the glia marker glutamine synthetase, show that IRAP is expressed in neurons. IRAP immunoreactive cells were observed in the hippocampal formation, cerebral cortex, thalamus, amygdala and, abundantly, hypothalamus. Double labeling experiments (IRAP and oxytocin/neurophysin 1, IRAP with vasopressin/neurophysin 2) revealed that IRAP is present in oxytocinergic and in vasopressinergic neurons. In schizophrenia patients, the numerical density of IRAP-expressing neurons in the paraventricular and the suprachiasmatic nuclei is significantly reduced, which might be associated with the reduction in neurophysin-containing neurons in these nuclei in schizophrenia. The pathophysiological role of lowered hypothalamic IRAP expression in schizophrenia remains to be established.


Assuntos
Cistinil Aminopeptidase/metabolismo , Hipotálamo/enzimologia , Hipotálamo/patologia , Neurônios/enzimologia , Neuro-Hipófise/metabolismo , Esquizofrenia/patologia , Idoso , Autopsia , Doença Crônica , Feminino , Glutamato-Amônia Ligase/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Neurofisinas/metabolismo , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Supraquiasmático/patologia , Vasopressinas/metabolismo
8.
FASEB J ; 31(1): 388-399, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733449

RESUMO

The suprachiasmatic nucleus (SCN)-often referred to as the master circadian clock-is essential in generating physiologic rhythms and orchestrating synchrony among circadian clocks. This study tested the hypothesis that periodic motivation induced by rhythmically pairing 2 reinforcing stimuli [methamphetamine (Meth) and running wheel (RW)] restores autonomous circadian activity in arrhythmic SCN-lesioned (SCNX) C3H/HeN mice. Sham-surgery and SCNX mice were treated with either Meth (1.2 mg/kg, i.p.) or vehicle in association, dissociation, or absence of an RW. Only the association of Meth treatment and restricted RW access successfully reestablished entrained circadian rhythms in mice with SCNX. RW-likely acting as a link between the circadian and reward systems-promotes circadian entrainment of activity. We conclude that a conditioned drug response is a powerful tool to entrain, drive, and restore circadian physiology. Furthermore, an RW should be recognized as a potent input signal in addition to the conventional use as an output signal.-Rawashdeh, O., Clough, S. J., Hudson, R. L., Dubocovich, M. L. Learned motivation drives circadian physiology in the absence of the master circadian clock.


Assuntos
Ritmo Circadiano/fisiologia , Aprendizagem/fisiologia , Motivação/fisiologia , Núcleo Supraquiasmático/patologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Ritmo Circadiano/efeitos dos fármacos , Masculino , Metanfetamina/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Motivação/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Núcleo Supraquiasmático/fisiologia
9.
Sci Rep ; 6: 36335, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27824104

RESUMO

Alzheimer's disease (AD) patients suffer sleep disorders and circadian rhythm disturbances (CRDs). The underlying mechanisms are incompletely understood, and treatments are lacking. In this study, we characterized the locomotor activity, clock gene expression, morphological degeneration and energy metabolism of suprachiasmatic nucleus (SCN), together with retinal light sensing, in ApoE-/- mice, a model for AD. Compared with the control C57BL/6J mice, ApoE-/- mice exhibited disordered circadian locomotor activity under dim light and constant darkness, with impaired re-entrainment to phase change schedules. Decreased retinal melanopsin expression, together with amyloidosis and tau deposition, was evident in ApoE-/- mice. Mitochondrial and synaptic deterioration, altered SIRT1-mediated energy metabolism and clock gene expression were also observed in ApoE-/- SCN. Supplementation with fat or ketone bodies but not glucose, or intraperitoneal administration of nicotinamide, restored the locomotor rhythmicity and circadian expression of SIRT1 and clock genes, as well as reducing neurodegeneration. Taken together, ApoE deficiency induced degeneration and a significant disturbance in the SCN rhythmicity. Decline of retinal light sensing and SCN structural and metabolic deteriorations represented the major pathologies accounting for the CRDs in ApoE-/- mice. Our curative experiments may help develop future therapies to treat the CRDs and sleep disorders in AD patients.


Assuntos
Doença de Alzheimer/genética , Proteínas CLOCK/metabolismo , Ritmo Circadiano , Sirtuína 1/metabolismo , Núcleo Supraquiasmático/patologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Modelos Animais de Doenças , Metabolismo Energético , Luz , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Mitocôndrias/patologia , Estimulação Luminosa , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiopatologia
10.
Bone ; 84: 139-147, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26721736

RESUMO

The neuropeptide Y system is known to play an important role in the regulation of bone homeostasis and while the functions of its major receptors, Y1R and Y2R, in this process have become clearer, the contributions of other Y-receptors, like the y6 receptor (y6R), are unknown. Y6R expression is restricted to the suprachiasmatic nucleus (SCN) of the hypothalamus, an area known to regulate circadian rhythms, and the testis. Here we show that lack of y6R signalling, results in significant reduction in bone mass, but no changes in bone length. Male and female y6R knockout (KO) mice display reduced cortical and cancellous bone volume in axial and appendicular bones. Mechanistically, the reduction in cancellous bone is the result of an uncoupling of bone remodelling, leading to an increase in osteoclast surface and number, and a reduction in osteoblast number, osteoid surface, mineralizing surface and bone formation rate. y6R KO mice displayed increased numbers of osteoclast precursors and produced greater numbers of osteoclasts in RANKL-treated cultures. They also produced fewer CFU-ALP osteoblast precursors in the marrow and showed reduced mineralization in primary osteoblastic cultures, as well as reduced expression for the osteoblast lineage marker, alkaline phosphatase, in bone isolates. The almost exclusive location of y6Rs in the hypothalamus suggests a critical role of central neuronal pathways controlling this uncoupling of bone remodelling which is in line with known actions or other Y-receptors in the brain. In conclusion, y6R signalling is required for maintenance of bone mass, with loss of y6R uncoupling bone remodelling and resulting in a negative bone balance. This study expands the scope of hypothalamic regulation of bone, highlighting the importance for neural/endocrine coordination and their marked effect upon skeletal homeostasis.


Assuntos
Reabsorção Óssea/metabolismo , Osteogênese , Receptores de Neuropeptídeo Y/metabolismo , Núcleo Supraquiasmático/metabolismo , Envelhecimento/metabolismo , Animais , Medula Óssea/metabolismo , Reabsorção Óssea/patologia , Calcificação Fisiológica , Contagem de Células , Diferenciação Celular , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/patologia , Osteócitos/metabolismo , Osteócitos/patologia , Osteogênese/genética , Receptores de Neuropeptídeo Y/deficiência , Receptores de Neuropeptídeo Y/genética , Transdução de Sinais , Núcleo Supraquiasmático/patologia
11.
J Biol Rhythms ; 31(2): 170-81, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26801650

RESUMO

The ventral subparaventricular zone (vSPVZ) receives direct retinal input and influences the daily patterning of activity in rodents, making it a likely candidate for the mediation of acute behavioral responses to light (i.e., masking). We performed chemical lesions aimed at the vSPVZ of diurnal grass rats (Arvicanthis niloticus) using N-methyl-D,L-aspartic acid (NMA), a glutamate agonist. Following NMA lesions, we placed grass rats in various lighting conditions (e.g., 12:12 light-dark, constant dark, constant light); presented a series of light pulses at circadian times (CT) 6, 14, 18, and 22; and placed them in a 7-h ultradian cycle to assess behavioral masking. Extensive bilateral lesions of the vSPVZ disrupted the expression of circadian rhythms of activity and abolished the circadian modulation of masking responses to light, without affecting light-induced masking behavior per se. We also found that in diurnal grass rats, NMA was capable of destroying not only neurons of the vSPVZ but also those of the suprachiasmatic nucleus (SCN), even though excitotoxins have been ineffective at destroying cells within the SCN of nocturnal rodents. The vulnerability of the grass rat's SCN to NMA toxicity raises the possibility of a difference in density of receptors for glutamate between nocturnal and diurnal species. In cases in which damage extended to the SCN, masking responses to light were present and similar to those displayed by animals with damage restricted to the vSPVZ. Thus, extensive bilateral lesions of the SCN and vSPVZ disrupted the expression of circadian rhythms without affecting acute responses to light in a diurnal species. Our present and previous results suggest that retinorecipient brain areas other than the SCN or vSPVZ, such as the intergeniculate leaflet or olivary pretectal nucleus, may be responsible for the mediation of masking responses to light in the diurnal grass rat.


Assuntos
Encéfalo/fisiologia , Ritmo Circadiano , Luz , Núcleo Supraquiasmático/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fotoperíodo , Proteínas Proto-Oncogênicas c-fos , Ratos , Núcleo Supraquiasmático/efeitos dos fármacos , Núcleo Supraquiasmático/patologia
12.
Neurobiol Dis ; 73: 366-76, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25448764

RESUMO

Despite advances in promoting axonal regeneration after adult central nervous system injury, elicitation of a large number of lesion-passing axons reform active synaptic connections with natural target neurons remains limited. By deleting both Pten and Socs3 in retinal ganglion cells, we report that optic nerve axons after prechiasm lesion robustly reinnervate the hypothalamus, form new synapses with neurons in the suprachiasmatic nucleus (SCN), and re-integrate with the existing circuitry. Photic or electric stimulation of the retinal axons induces neuronal response in SCN. However both the innervation pattern and evoked responses are not completely restored by the regenerating axons, suggesting that combining with other strategies is necessary to overcome the defective rewiring. Our results support that boosting the intrinsic growth capacity in injured neurons promotes axonal reinnervation and rewiring.


Assuntos
Axônios/fisiologia , Hipotálamo/fisiologia , Regeneração Nervosa/fisiologia , Nervo Óptico/fisiologia , PTEN Fosfo-Hidrolase , Células Ganglionares da Retina/fisiologia , Proteínas Supressoras da Sinalização de Citocina , Núcleo Supraquiasmático/fisiologia , Sinapses/fisiologia , Animais , Axônios/patologia , Hipotálamo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nervo Óptico/patologia , PTEN Fosfo-Hidrolase/genética , Células Ganglionares da Retina/patologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Núcleo Supraquiasmático/patologia , Sinapses/patologia
13.
PLoS One ; 9(5): e97651, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24830778

RESUMO

The orexigenic gut-brain peptide, ghrelin and its G-protein coupled receptor, the growth hormone secretagogue receptor 1a (GHS-R1A) are pivotal regulators of hypothalamic feeding centers and reward processing neuronal circuits of the brain. These systems operate in a cooperative manner and receive a wide array of neuronal hormone/transmitter messages and metabolic signals. Functional magnetic resonance imaging was employed in the current study to map BOLD responses to ghrelin in different brain regions with special reference on homeostatic and hedonic regulatory centers of energy balance. Experimental groups involved male, ovariectomized female and ovariectomized estradiol-replaced rats. Putative modulation of ghrelin signaling by endocannabinoids was also studied. Ghrelin-evoked effects were calculated as mean of the BOLD responses 30 minutes after administration. In the male rat, ghrelin evoked a slowly decreasing BOLD response in all studied regions of interest (ROI) within the limbic system. This effect was antagonized by pretreatment with GHS-R1A antagonist JMV2959. The comparison of ghrelin effects in the presence or absence of JMV2959 in individual ROIs revealed significant changes in the prefrontal cortex, nucleus accumbens of the telencephalon, and also within hypothalamic centers like the lateral hypothalamus, ventromedial nucleus, paraventricular nucleus and suprachiasmatic nucleus. In the female rat, the ghrelin effects were almost identical to those observed in males. Ovariectomy and chronic estradiol replacement had no effect on the BOLD response. Inhibition of the endocannabinoid signaling by rimonabant significantly attenuated the response of the nucleus accumbens and septum. In summary, ghrelin can modulate hypothalamic and mesolimbic structures controlling energy balance in both sexes. The endocannabinoid signaling system contributes to the manifestation of ghrelin's BOLD effect in a region specific manner. In females, the estradiol milieu does not influence the BOLD response to ghrelin.


Assuntos
Encéfalo/patologia , Regulação da Expressão Gênica , Grelina/metabolismo , Homeostase , Imageamento por Ressonância Magnética , Animais , Encéfalo/metabolismo , Estradiol/metabolismo , Comportamento Alimentar , Feminino , Hipotálamo/metabolismo , Hipotálamo/patologia , Sistema Límbico/fisiologia , Masculino , Núcleo Accumbens/patologia , Núcleo Hipotalâmico Paraventricular/patologia , Córtex Pré-Frontal/patologia , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/metabolismo , Recompensa , Transdução de Sinais , Núcleo Supraquiasmático/patologia , Fatores de Tempo
14.
Cell Metab ; 19(1): 58-72, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24411939

RESUMO

Y-receptors control energy homeostasis, but the role of Npy6 receptors (Npy6r) is largely unknown. Young Npy6r-deficient (Npy6r(-/-)) mice have reduced body weight, lean mass, and adiposity, while older and high-fat-fed Npy6r(-/-) mice have low lean mass with increased adiposity. Npy6r(-/-) mice showed reduced hypothalamic growth hormone releasing hormone (Ghrh) expression and serum insulin-like growth factor-1 (IGF-1) levels relative to WT. This is likely due to impaired vasoactive intestinal peptide (VIP) signaling in the suprachiasmatic nucleus (SCN), where we found Npy6r coexpressed in VIP neurons. Peripheral administration of pancreatic polypeptide (PP) increased Fos expression in the SCN, increased energy expenditure, and reduced food intake in WT, but not Npy6r(-/-), mice. Moreover, intraperitoneal (i.p.) PP injection increased hypothalamic Ghrh mRNA expression and serum IGF-1 levels in WT, but not Npy6r(-/-), mice, an effect blocked by intracerebroventricular (i.c.v.) Vasoactive Intestinal Peptide (VPAC) receptors antagonism. Thus, PP-initiated signaling through Npy6r in VIP neurons regulates the growth hormone axis and body composition.


Assuntos
Metabolismo Energético , Homeostase , Polipeptídeo Pancreático/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais , Núcleo Supraquiasmático/metabolismo , Adiposidade , Animais , Peso Corporal , Corticosterona/metabolismo , Dieta , Comportamento Alimentar , Fertilidade , Fator de Crescimento Insulin-Like I/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/patologia , Receptores dos Hormônios Gastrointestinais/deficiência , Receptores de Neuropeptídeo Y/deficiência , Núcleo Supraquiasmático/patologia , Magreza/sangue , Magreza/patologia , Peptídeo Intestinal Vasoativo/metabolismo
15.
PLoS One ; 8(11): e80029, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24244599

RESUMO

Dysregulation of circadian rhythmicity is identified as a key factor in disease pathogenesis. Circadian rhythmicity is controlled at both a transcriptional and post-transcriptional level suggesting the role of microRNA (miRNA) and double-stranded RNA (dsRNA) in this process. Endonuclease Dicer controls miRNA and dsRNA processing, however the role of Dicer in circadian regulation is not known. Here we demonstrate robust diurnal oscillations of Dicer expression in central and peripheral clock control systems including suprachiasmatic nucleolus (SCN), retina, liver, and bone marrow (BM). The Dicer oscillations were either reduced or phase shifted with aging and Type 2 diabetes. The decrease and phase shift of Dicer expression was associated with a similar decrease and phase shift of miRNAs 146a and 125a-5p and with an increase in toxic Alu RNA. Restoring Dicer levels and the diurnal patterns of Dicer-controlled miRNA and RNA expression may provide new therapeutic strategies for metabolic disease and aging-associated complications.


Assuntos
Envelhecimento/genética , RNA Helicases DEAD-box/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , RNA Mensageiro/genética , Ribonuclease III/genética , Adulto , Idoso , Envelhecimento/metabolismo , Envelhecimento/patologia , Elementos Alu/genética , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Ritmo Circadiano/genética , RNA Helicases DEAD-box/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Mensageiro/metabolismo , Retina/metabolismo , Retina/patologia , Ribonuclease III/metabolismo , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia
16.
Proc Natl Acad Sci U S A ; 108(14): 5813-8, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21402951

RESUMO

Food anticipatory behavior (FAA) is induced by limiting access to food for a few hours daily. Animals anticipate this scheduled meal event even without the suprachiasmatic nucleus (SCN), the biological clock. Consequently, a food-entrained oscillator has been proposed to be responsible for meal time estimation. Recent studies suggested the dorsomedial hypothalamus (DMH) as the site for this food-entrained oscillator, which has led to considerable controversy in the literature. Herein we demonstrate by means of c-Fos immunohistochemistry that the neuronal activity of the suprachiasmatic nucleus (SCN), which signals the rest phase in nocturnal animals, is reduced when animals anticipate the scheduled food and, simultaneously, neuronal activity within the DMH increases. Using retrograde tracing and confocal analysis, we show that inhibition of SCN neuronal activity is the consequence of activation of GABA-containing neurons in the DMH that project to the SCN. Next, we show that DMH lesions result in a loss or diminution of FAA, simultaneous with increased activity in the SCN. A subsequent lesion of the SCN restored FAA. We conclude that in intact animals, FAA may only occur when the DMH inhibits the activity of the SCN, thus permitting locomotor activity. As a result, FAA originates from a neuronal network comprising an interaction between the DMH and SCN. Moreover, this study shows that the DMH-SCN interaction may serve as an intrahypothalamic system to gate activity instead of rest overriding circadian predetermined temporal patterns.


Assuntos
Antecipação Psicológica/fisiologia , Núcleo Hipotalâmico Dorsomedial/fisiologia , Alimentos , Modelos Neurológicos , Núcleo Supraquiasmático/fisiologia , Animais , Núcleo Hipotalâmico Dorsomedial/patologia , Imuno-Histoquímica , Ácido Caínico , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Núcleo Supraquiasmático/patologia
17.
J Biol Rhythms ; 25(6): 432-41, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21135159

RESUMO

In mammals, a pacemaker in the suprachiasmatic nucleus (SCN) is thought to be required for behavioral, physiological, and molecular circadian rhythms. However, there is considerable evidence that temporal food restriction (restricted feedisng [RF]) and chronic methamphetamine (MA) can drive circadian rhythms of locomotor activity, body temperature, and endocrine function in the absence of SCN. This indicates the existence of extra-SCN pacemakers: the Food Entrainable Oscillator (FEO) and Methamphetamine Sensitive Circadian Oscillator (MASCO). Here, we show that these extra-SCN pacemakers control the phases of peripheral oscillators in intact as well as in SCN-ablated PER2::LUC mice. MA administration shifted the phases of SCN, cornea, pineal, pituitary, kidney, and salivary glands in intact animals. When the SCN was ablated, disrupted phase relationships among peripheral oscillators were reinstated by MA treatment. When intact animals were subjected to restricted feeding, the phases of cornea, pineal, kidney, salivary gland, lung, and liver were shifted. In SCN-lesioned restricted-fed mice, phases of all of the tissues shifted such that they aligned with the time of the meal. Taken together, these data show that FEO and MASCO are strong circadian pacemakers able to regulate the phases of peripheral oscillators.


Assuntos
Ritmo Circadiano/fisiologia , Atividade Motora/fisiologia , Proteínas Circadianas Period/fisiologia , Núcleo Supraquiasmático/fisiopatologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Córnea/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Rim/fisiologia , Luciferases/genética , Luciferases/metabolismo , Masculino , Metanfetamina/farmacologia , Camundongos , Camundongos Transgênicos , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Glândula Pineal/fisiologia , Hipófise/fisiologia , Glândulas Salivares/fisiologia , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia
18.
Chronobiol Int ; 27(6): 1159-77, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20653448

RESUMO

Using a rodent model for neuropathology induced by human amyloid precursor protein, the present study tested the hypothesis that 24 h rest/activity rhythms deteriorate with age. A lifespan of rest/activity patterns was studied in transgenic Tg2576 mice and wild-type controls. Classic indices of circadian timekeeping, including onsets, offsets, and the duration of nighttime activity, were stable throughout the 96-week study. Analyses of ultradian bout activity revealed significant genotype and age-related changes in the duration and intensity of activity bouts, as well as amplitude of the 24 h rhythm. Tg2576 mice had more total activity counts, fewer bouts/24 h, more counts/bout, and longer bout time than wild-type controls. Amyloid deposits and plaques were solely found in specific cortex regions in aged postmortem Tg2576 mice, but were not evident in the hypothalamus or suprachiasmatic nucleus; this neuropathology was absent from brains of wild-type controls. These findings suggest that amyloidosis of the Tg2576 mouse exerts little influence on timing of locomotor activity in the circadian domain but significantly alters the temporal structure of ultradian activity.


Assuntos
Ciclos de Atividade/fisiologia , Envelhecimento/fisiologia , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/fisiologia , Ritmo Circadiano/fisiologia , Ciclos de Atividade/genética , Envelhecimento/genética , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Ritmo Circadiano/genética , Modelos Animais de Doenças , Humanos , Longevidade/genética , Longevidade/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Núcleo Supraquiasmático/patologia , Núcleo Supraquiasmático/fisiopatologia
19.
Usp Fiziol Nauk ; 39(4): 40-65, 2008.
Artigo em Russo | MEDLINE | ID: mdl-19241804

RESUMO

The current data on the role of pineal gland in the regulation of biorhythms of an organism are critically reviewed. The changes of structure and functional status of the pineal gland and of the hypothalamic suprachyasmatic nucleus during normal aging and at age-associated pathology are presented as well. The particular attention are given to effects of light/dark regimen disturbances and shift work on the process of aging and development of pathology, cancer including. At last, the available data effects of exogenous melatonin on life span and tumorigenesis were observed.


Assuntos
Envelhecimento/fisiologia , Melatonina/metabolismo , Periodicidade , Glândula Pineal/fisiologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Proteínas CLOCK , Humanos , Luz , Melatonina/farmacologia , Neoplasias/etiologia , Glândula Pineal/anatomia & histologia , Glândula Pineal/patologia , Núcleo Supraquiasmático/anatomia & histologia , Núcleo Supraquiasmático/patologia , Transativadores/genética
20.
Neurobiol Aging ; 28(8): 1239-47, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16837102

RESUMO

The pineal hormone melatonin is involved in the regulation of circadian rhythms and feeds back to the central biological clock, the hypothalamic suprachiasmatic nucleus (SCN) via melatonin receptors. Supplementary melatonin is considered to be a potential treatment for aging and Alzheimer's disease (AD)-related circadian disorders. Here we investigated by immunocytochemistry the alterations of the MT1 melatonin receptor, the neuropeptides vasopressin (AVP) and vasoactive intestinal peptide (VIP) in the SCN during aging and AD. We found that the number and density of AVP/VIP-expressing neurons in the SCN did not change, but the number and density of MT1-expressing neurons in the SCN were decreased in aged controls compared to young controls. Furthermore, both MT1-expressing neurons and AVP/VIP-expressing neurons were strongly diminished in the last neuropathological stages of AD (Braak stages V-VI), but not in the earliest stages (Braak stages I-II), compared to aged controls (Braak stage 0). Our study suggests that the MT1-mediated effects of melatonin on the SCN are disturbed during aging and even more so in late stage AD, which may contribute to the clinical circadian disorders and to the efficacy of therapeutic melatonin administration under these conditions.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/patologia , Regulação da Expressão Gênica/fisiologia , Receptor MT1 de Melatonina/metabolismo , Núcleo Supraquiasmático/metabolismo , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Mudanças Depois da Morte , Receptor MT1 de Melatonina/genética , Estatísticas não Paramétricas , Núcleo Supraquiasmático/patologia , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/metabolismo , Vasopressinas/genética , Vasopressinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA