Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neuropharmacology ; 257: 110050, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38914372

RESUMO

Animals inhabiting temperate and high latitudes undergo drastic seasonal changes in energy storage, facilitated by changes in food intake and body mass. Those seasonal changes in the animal's biology are not mere consequences of environmental energy availability but are anticipatory responses to the energetic requirements of the upcoming season and are actively timed by tracking the annual progression in photoperiod. In this review, we discuss how photoperiod is used to control energy balance seasonally and how this is distinct from energy homeostasis. Most notably, we suggest that photoperiodic control of food intake and body mass does not originate from the arcuate nucleus, as for homeostatic appetite control, but is rather to be found in hypothalamic tanycytes. Tanycytes are specialized ependymal cells lining the third ventricle, which can sense metabolites from the cerebrospinal fluid (e.g. glucose) and can control access of circulating signals to the brain. They are also essential in conveying time-of-year information by integrating photoperiod and altering hypothalamic thyroid metabolism, a feature that is conserved in seasonal vertebrates and connects to seasonal breeding and metabolism. We also discuss how homeostatic feedback signals are handled during times of rapid energetic transitions. Studies on leptin in seasonal mammals suggest a seasonal shift in central sensitivity and blood-brain transport, which might be facilitated by tanycytes.

2.
J Exp Biol ; 227(10)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38690647

RESUMO

Hibernation is an extreme state of seasonal energy conservation, reducing metabolic rate to as little as 1% of the active state. During the hibernation season, many species of hibernating mammals cycle repeatedly between the active (aroused) and hibernating (torpid) states (T-A cycling), using brown adipose tissue (BAT) to drive cyclical rewarming. The regulatory mechanisms controlling this process remain undefined but are presumed to involve thermoregulatory centres in the hypothalamus. Here, we used the golden hamster (Mesocricetus auratus), and high-resolution monitoring of BAT, core body temperature and ventilation rate, to sample at precisely defined phases of the T-A cycle. Using c-fos as a marker of cellular activity, we show that although the dorsomedial hypothalamus is active during torpor entry, neither it nor the pre-optic area shows any significant changes during the earliest stages of spontaneous arousal. Contrastingly, in three non-neuronal sites previously linked to control of metabolic physiology over seasonal and daily time scales - the choroid plexus, pars tuberalis and third ventricle tanycytes - peak c-fos expression is seen at arousal initiation. We suggest that through their sensitivity to factors in the blood or cerebrospinal fluid, these sites may mediate metabolic feedback-based initiation of the spontaneous arousal process.


Assuntos
Nível de Alerta , Plexo Corióideo , Células Ependimogliais , Hibernação , Proteínas Proto-Oncogênicas c-fos , Torpor , Animais , Proteínas Proto-Oncogênicas c-fos/metabolismo , Nível de Alerta/fisiologia , Torpor/fisiologia , Hibernação/fisiologia , Células Ependimogliais/metabolismo , Células Ependimogliais/fisiologia , Plexo Corióideo/metabolismo , Plexo Corióideo/fisiologia , Mesocricetus , Masculino , Tecido Adiposo Marrom/fisiologia , Tecido Adiposo Marrom/metabolismo , Cricetinae
3.
Curr Biol ; 34(3): 632-640.e6, 2024 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-38218183

RESUMO

In mammals, maternal photoperiodic programming (MPP) provides a means whereby juvenile development can be matched to forthcoming seasonal environmental conditions.1,2,3,4 This phenomenon is driven by in utero effects of maternal melatonin5,6,7 on the production of thyrotropin (TSH) in the fetal pars tuberalis (PT) and consequent TSH receptor-mediated effects on tanycytes lining the 3rd ventricle of the mediobasal hypothalamus (MBH).8,9,10 Here we use LASER capture microdissection and transcriptomic profiling to show that TSH-dependent MPP controls the attributes of the ependymal region of the MBH in juvenile animals. In Siberian hamster pups gestated and raised on a long photoperiod (LP) and thereby committed to a fast trajectory for growth and reproductive maturation, the ependymal region is enriched for tanycytes bearing sensory cilia and receptors implicated in metabolic sensing. Contrastingly, in pups gestated and raised on short photoperiod (SP) and therefore following an over-wintering developmental trajectory with delayed sexual maturation, the ependymal region has fewer sensory tanycytes. Post-weaning transfer of SP-gestated pups to an intermediate photoperiod (IP), which accelerates reproductive maturation, results in a pronounced shift toward a ciliated tanycytic profile and formation of tanycytic processes. We suggest that tanycytic plasticity constitutes a mechanism to tailor metabolic development for extended survival in variable overwintering environments.


Assuntos
Células Ependimogliais , Melatonina , Cricetinae , Animais , Células Ependimogliais/metabolismo , Estações do Ano , Hipotálamo/metabolismo , Ritmo Circadiano , Phodopus/metabolismo , Fotoperíodo , Tireotropina/metabolismo
4.
J Biol Rhythms ; 38(6): 586-600, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37565646

RESUMO

Seasonal mammals register photoperiodic changes through the photoneuroendocrine system enabling them to time seasonal changes in growth, metabolism, and reproduction. To a varying extent, proximate environmental factors like ambient temperature (Ta) modulate timing of seasonal changes in physiology, conferring adaptive flexibility. While the molecular photoneuroendocrine pathway governing the seasonal responses is well defined, the mechanistic integration of nonphotoperiodic modulatory cues is poorly understood. Here, we explored the interaction between Ta and photoperiod in tundra voles, Microtus oeconomus, a boreal species in which the main impact of photoperiod is on postnatal somatic growth. We demonstrate that postweaning growth potential depends on both gestational and postweaning patterns of photoperiodic exposure, with the highest growth potential seen in voles experiencing short (8 h) gestational and long (16 h) postweaning photoperiods-corresponding to a spring growth program. Modulation by Ta was asymmetric: low Ta (10 °C) enhanced the growth potential of voles gestated on short photoperiods independent of postweaning photoperiod exposure, whereas in voles gestated on long photoperiods, showing a lower autumn-programmed growth potential, the effect of Ta was highly dependent on postweaning photoperiod. Analysis of the primary molecular elements involved in the expression of a neuroendocrine response to photoperiod, thyrotropin beta subunit (tshß) in the pars tuberalis, somatostatin (srif) in the arcuate nucleus, and type 2/3 deiodinase (dio2/dio3) in the mediobasal hypothalamus identified dio2 as the most Ta-sensitive gene across the study, showing increased expression at higher Ta, while higher Ta reduced somatostatin expression. Contrastingly dio3 and tshß were largely insensitive to Ta. Overall, these observations reveal a complex interplay between Ta and photoperiodic control of postnatal growth in M. oeconomus, and suggest that integration of Ta into the control of growth occurs downstream of the primary photoperiodic response cascade revealing potential adaptivity of small herbivores facing rising temperatures at high latitudes.


Assuntos
Ritmo Circadiano , Fotoperíodo , Animais , Estações do Ano , Temperatura , Arvicolinae , Somatostatina , Tundra
5.
ACS Appl Mater Interfaces ; 14(4): 5066-5079, 2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35041392

RESUMO

Microglia are the major innate immune cells in the brain and are essential for maintaining homeostasis in a neuronal microenvironment. Currently, a genetic tool to modify microglial gene expression in specific brain regions is not available. In this report, we introduce a tailor-designed method that uses lipid and polymer hybridized nanoparticles (LPNPs) for the local delivery of small interfering RNAs (siRNAs), allowing the silencing of specific microglial genes in the hypothalamus. Our physical characterization proved that this LPNP-siRNA was uniform and stable. We demonstrated that, due to their natural phagocytic behavior, microglial cells are the dominant cell type taking up these LPNPs in the hypothalamus of rats. We then tested the silencing efficiency of LPNPs carrying a cluster of differentiation molecule 11b (CD11b) or Toll-like receptor 4 (TLR4) siRNA using different in vivo and in vitro approaches. In cultured microglial cells treated with LPNP-CD11b siRNA or LPNP-TLR4 siRNA, we found a silencing efficiency at protein expression levels of 65 or 77%, respectively. In line with this finding, immunohistochemistry and western blotting results from in vivo experiments showed that LPNP-CD11b siRNA significantly inhibited microglial CD11b protein expression in the hypothalamus. Furthermore, following lipopolysaccharide (LPS) stimulation of cultured microglial cells, gene expression of the TLR4 downstream signaling component myeloid differentiation factor 88 and its associated cytokines was significantly inhibited in LPNP-TLR4 siRNA-treated microglial cells compared with cells treated with LPNP-scrambled siRNA. Finally, after LPNP-TLR4 siRNA injection into the rat hypothalamus, we observed a significant reduction in microglial activation in response to LPS compared with the control rats injected with LPNP-scrambled siRNA. Our results indicate that LPNP-siRNA is a promising tool to manipulate microglial activity locally in the brain and may serve as a prophylactic approach to prevent microglial dysfunction-associated diseases.


Assuntos
Portadores de Fármacos/química , Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Microglia/efeitos dos fármacos , Nanopartículas/química , RNA Interferente Pequeno/farmacologia , Animais , Antígeno CD11b/antagonistas & inibidores , Antígeno CD11b/genética , Lipídeos/química , Masculino , Poliésteres/química , Polietilenoglicóis/química , Ratos Wistar , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética
6.
J Neuroendocrinol ; 33(7): e12973, 2021 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-33960524

RESUMO

Kisspeptin (Kp) and (Arg)(Phe) related peptide 3 (RFRP-3) are two RF-amides acting in the hypothalamus to control reproduction. In the past 10 years, it has become clear that, apart from their role in reproductive physiology, both neuropeptides are also involved in the control of food intake, as well as glucose and energy metabolism. To investigate further the neural mechanisms responsible for these metabolic actions, we assessed the effect of acute i.c.v. administration of Kp or RFRP-3 in ad lib. fed male Wistar rats on feeding behaviour, glucose and energy metabolism, circulating hormones (luteinising hormone, testosterone, insulin and corticosterone) and hypothalamic neuronal activity. Kp increased plasma testosterone levels, had an anorexigenic effect and increased lipid catabolism, as attested by a decreased respiratory exchange ratio (RER). RFRP-3 also increased plasma testosterone levels but did not modify food intake or energy metabolism. Both RF-amides increased endogenous glucose production, yet with no change in plasma glucose levels, suggesting that these peptides provoke not only a release of hepatic glucose, but also a change in glucose utilisation. Finally, plasma insulin and corticosterone levels did not change after the RF-amide treatment. The Kp effects were associated with an increased c-Fos expression in the median preoptic area and a reduction in pro-opiomelanocortin immunostaining in the arcuate nucleus. No effects on neuronal activation were found for RFRP-3. Our results provide further evidence that Kp is not only a very potent hypothalamic activator of reproduction, but also part of the hypothalamic circuit controlling energy metabolism.

7.
J Neuroendocrinol ; 32(5): e12845, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32291844

RESUMO

In addition to its regulatory role in luteinising hormone secretion, Rfamide-related peptide 3 (RFRP3) has also been reported to modulate food intake in several mammalian species. Djungarian hamsters (Phodopus sungorus), similar to other seasonal mammals, display a remarkable inhibition of RFRP3 expression in winter short-day conditions, associated with decreased food intake and bodyweight. This species is therefore a valuable model for assessing whether RFRP3 might be involved in the seasonal control of feeding behaviour and investigating its possible brain targets. We found that, although both male and female animals exhibit the same robust reduction in Rfrp expression in short- (SD) compared to long-day (LD) conditions, acute central administration of RFRP3 displays sex-dependent effects on food intake. RFRP3 increased food intake in female hamsters in SD or in LD dioestrus, but not in LD pro-oestrus, indicating that the orexigenic effect of RFRP3 is observed in conditions of low circulating oestradiol levels. In male hamsters, food intake was not changed by acute injections of RFRP3, regardless of whether animals were in SD or LD conditions. Analysing the gene expression of various metabolic neuropeptides in the brain of RFRP3-injected Djungarian hamsters revealed that Npy expression was increased in female but not in male animals. The present study suggests that, in Djungarian hamsters, RFRP3 exhibits a sex-dependent orexigenic effect possibly by inducing increased Npy expression.


Assuntos
Neuropeptídeos , Phodopus , Animais , Cricetinae , Ingestão de Alimentos , Feminino , Masculino , Neuropeptídeos/farmacologia , Phodopus/fisiologia , Fotoperíodo , Estações do Ano
8.
J Neuroendocrinol ; 31(4): e12710, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30887598

RESUMO

Many animals exhibit remarkable metabolic and reproductive adaptations to seasonal changes in their environment. When day length shortens, Djungarian hamsters (Phodopus sungorus) reduce their body weight and inhibit their reproductive activity, whereas the opposite occurs in springtime. These physiological adaptations are considered to depend on photoperiodic changes in hypothalamic genes encoding the peptides kisspeptin (Kp) and RFamide-related peptide 3 (RFRP3) for the control of reproduction, as well as pro-opiomelanocortin and somatostatin for metabolic regulation. The present study investigates the effect of Kp and RFRP3 on long-term body weight regulation, aiming to establish whether metabolic and reproductive hypothalamic networks may interact during adaptation to seasonal physiology. We found that chronic central administration of both Kp and RFRP3 in short photoperiod-adapted male Djungarian hamsters increased body weight, although via different pathways. The effect of Kp was dependent on testicular activity because castration prevented the body weight increase and was associated with an increase in pro-opiomelanocortin and neuropeptide Y expression. On the other hand, the orexigenic effect of RFRP3 was associated with an increase in circulating insulin and leptin levels, although it had no effect on any of the hypothalamic metabolic genes investigated, and did not change circulating levels of sex steroids. Notably, neither Kp, nor RFRP3 altered female hamster metabolic parameters. Thus, using a rodent model exhibiting seasonal changes in reproduction and metabolism, the present study demonstrates that, in addition to its role in the central control of reproduction, Kp also participates in body weight control in a sex-dependent manner via an anabolic action of testosterone. Conversely, RFRP3 affects body weight control in males mostly by acting on adiposity, with no overt effect on the reproductive system in both sexes.


Assuntos
Peso Corporal/efeitos dos fármacos , Kisspeptinas/farmacologia , Neuropeptídeos/farmacologia , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/fisiologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Insulina/sangue , Leptina/sangue , Masculino , Neuropeptídeo Y/genética , Phodopus/fisiologia , Fotoperíodo , Pró-Opiomelanocortina/genética , Reprodução/fisiologia , Estações do Ano , Fatores Sexuais , Testículo/fisiologia
9.
J Endocrinol ; 235(3): 167-178, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28851750

RESUMO

Circadian disruption is associated with metabolic disturbances such as hepatic steatosis (HS), obesity and type 2 diabetes. We hypothesized that HS, resulting from constant light (LL) exposure is due to an inconsistency between signals related to food intake and endocrine-driven suprachiasmatic nucleus (SCN) outputs. Indeed, exposing rats to LL induced locomotor, food intake and hormone arrhythmicity together with the development of HS. We investigated whether providing temporal signals such as 12-h food availability or driving a corticosterone plus melatonin rhythm could restore rhythmicity and prevent the metabolic disturbances under LL conditions in male rats. Discrete metabolic improvements under these separate treatments stimulated us to investigate whether the combination of hormone treatment together with mealtime restriction (12-h food during four weeks) could prevent the metabolic alterations. LL exposed arrhythmic rats, received daily administration of corticosterone (2.5 µg/kg) and melatonin (2.5 mg/kg) in synchrony or out of synchrony with their 12-h meal. HS and other metabolic alterations were importantly ameliorated in LL-exposed rats receiving hormonal treatment in synchrony with 12-h restricted mealtime, while treatment out of phase with meal time did not. Interestingly, liver bile acids, a major indication for HS, were only normalized when animals received hormones in synchrony with food indicating that disrupted bile acid metabolism might be an important mechanism for the HS induction under LL conditions. We conclude that food-elicited signals, as well as hormonal signals, are necessary for liver synchronization and that HS arises when there is conflict between food intake and the normal pattern of melatonin and corticosterone.


Assuntos
Transtornos Cronobiológicos/complicações , Corticosterona/administração & dosagem , Fígado Gorduroso/etiologia , Métodos de Alimentação , Melatonina/administração & dosagem , Núcleo Supraquiasmático/fisiopatologia , Adiposidade/efeitos dos fármacos , Animais , Transtornos Cronobiológicos/fisiopatologia , Transtornos Cronobiológicos/prevenção & controle , Fígado Gorduroso/metabolismo , Fígado Gorduroso/prevenção & controle , Transtornos do Metabolismo de Glucose/etiologia , Transtornos do Metabolismo de Glucose/prevenção & controle , Luz/efeitos adversos , Masculino , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA