Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neural Regen Res ; 19(9): 1919-1928, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38227516

RESUMO

Over the past century, age-related diseases, such as cancer, type-2 diabetes, obesity, and mental illness, have shown a significant increase, negatively impacting overall quality of life. Studies on aged animal models have unveiled a progressive discoordination at multiple regulatory levels, including transcriptional, translational, and post-translational processes, resulting from cellular stress and circadian derangements. The circadian clock emerges as a key regulator, sustaining physiological homeostasis and promoting healthy aging through timely molecular coordination of pivotal cellular processes, such as stem-cell function, cellular stress responses, and inter-tissue communication, which become disrupted during aging. Given the crucial role of hypothalamic circuits in regulating organismal physiology, metabolic control, sleep homeostasis, and circadian rhythms, and their dependence on these processes, strategies aimed at enhancing hypothalamic and circadian function, including pharmacological and non-pharmacological approaches, offer systemic benefits for healthy aging. Intranasal brain-directed drug administration represents a promising avenue for effectively targeting specific brain regions, like the hypothalamus, while reducing side effects associated with systemic drug delivery, thereby presenting new therapeutic possibilities for diverse age-related conditions.

2.
Nat Commun ; 14(1): 1685, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-36973248

RESUMO

The circadian clock is an endogenous time-tracking system that anticipates daily environmental changes. Misalignment of the clock can cause obesity, which is accompanied by reduced levels of the clock-controlled, rhythmic metabolite NAD+. Increasing NAD+ is becoming a therapy for metabolic dysfunction; however, the impact of daily NAD+ fluctuations remains unknown. Here, we demonstrate that time-of-day determines the efficacy of NAD+ treatment for diet-induced metabolic disease in mice. Increasing NAD+ prior to the active phase in obese male mice ameliorated metabolic markers including body weight, glucose and insulin tolerance, hepatic inflammation and nutrient sensing pathways. However, raising NAD+ immediately before the rest phase selectively compromised these responses. Remarkably, timed NAD+ adjusted circadian oscillations of the liver clock until completely inverting its oscillatory phase when increased just before the rest period, resulting in misaligned molecular and behavioral rhythms in male and female mice. Our findings unveil the time-of-day dependence of NAD+-based therapies and support a chronobiology-based approach.


Assuntos
Relógios Circadianos , Doenças Metabólicas , Camundongos , Masculino , Feminino , Animais , Ritmo Circadiano/fisiologia , NAD/metabolismo , Dieta , Doenças Metabólicas/metabolismo , Fígado/metabolismo
3.
Glia ; 71(7): 1626-1647, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36919670

RESUMO

Hypothalamic circuits compute systemic information to control metabolism. Astrocytes residing within the hypothalamus directly sense nutrients and hormones, integrating metabolic information, and modulating neuronal responses. Nevertheless, the role of the astrocytic circadian clock on the control of energy balance remains unclear. We used mice with a targeted ablation of the core-clock gene Bmal1 within Gfap-expressing astrocytes to gain insight on the role played by this transcription factor in astrocytes. While this mutation does not substantially affect the phenotype in mice fed normo-caloric diet, under high-fat diet we unmasked a thermogenic phenotype consisting of increased energy expenditure, and catabolism in brown adipose and overall metabolic improvement consisting of better glycemia control, and body composition. Transcriptomic analysis in the ventromedial hypothalamus revealed an enhanced response to moderate cellular stress, including ER-stress response, unfolded protein response and autophagy. We identified Xbp1 and Atf1 as two key transcription factors enhancing cellular stress responses. Therefore, we unveiled a previously unknown role of the astrocytic circadian clock modulating energy balance through the regulation of cellular stress responses within the VMH.


Assuntos
Relógios Circadianos , Camundongos , Animais , Relógios Circadianos/genética , Astrócitos/metabolismo , Hipotálamo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Metabolismo Energético/genética
4.
Neuropsychopharmacology ; 47(4): 805-816, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34837078

RESUMO

A growing number of epidemiological and experimental studies has established that circadian disruption is strongly associated with psychiatric disorders, including major depressive disorder (MDD). This association is becoming increasingly relevant considering that modern lifestyles, social zeitgebers (time cues) and genetic variants contribute to disrupting circadian rhythms that may lead to psychiatric disorders. Circadian abnormalities associated with MDD include dysregulated rhythms of sleep, temperature, hormonal secretions, and mood which are modulated by the molecular clock. Rapid-acting antidepressants such as subanesthetic ketamine and sleep deprivation therapy can improve symptoms within 24 h in a subset of depressed patients, in striking contrast to conventional treatments, which generally require weeks for a full clinical response. Importantly, animal data show that sleep deprivation and ketamine have overlapping effects on clock gene expression. Furthermore, emerging data implicate the circadian system as a critical component involved in rapid antidepressant responses via several intracellular signaling pathways such as GSK3ß, mTOR, MAPK, and NOTCH to initiate synaptic plasticity. Future research on the relationship between depression and the circadian clock may contribute to the development of novel therapeutic strategies for depression-like symptoms. In this review we summarize recent evidence describing: (1) how the circadian clock is implicated in depression, (2) how clock genes may contribute to fast-acting antidepressants, and (3) the mechanistic links between the clock genes driving circadian rhythms and neuroplasticity.


Assuntos
Relógios Circadianos , Transtorno Depressivo Maior , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Ritmo Circadiano/fisiologia , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/genética , Humanos , Privação do Sono/genética
5.
J Neurol Neuromedicine ; 3(1): 21-25, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29682634

RESUMO

Daily rhythms in behavior and physiology are coordinated by an endogenous clock located in the suprachiasmatic nucleus (SCN) of the hypothalamus. This central pacemaker also relays day length information to allow for seasonal adaptation, a process for which melatonin signaling is essential. How the SCN encodes day length is not fully understood. MicroRNAs (miRNAs) are small, non-coding RNAs that regulate gene expression by directing target mRNAs for degradation or translational repression. The miR-132/212 cluster plays a key role in facilitating neuronal plasticity, and miR-132 has been shown previously to modulate resetting of the central clock. A recent study from our group showed that miR-132/212 in mice is required for optimal adaptation to seasons and non-24-hour light/dark cycles through regulation of its target gene, methyl CpG-binding protein (MeCP2), in the SCN and dendritic spine density of SCN neurons. Furthermore, in the seasonal rodent Mesocricetus auratus (Syrian hamster), adaptation to short photoperiods is accompanied by structural plasticity in the SCN independently of melatonin signaling, thus further supporting a key role for SCN structural and, in turn, functional plasticity in the coding of day length. In this commentary, we discuss our recent findings in context of what is known about day length encoding by the SCN, and propose future directions.

6.
Cell Rep ; 19(3): 505-520, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423315

RESUMO

The central circadian pacemaker, the suprachiasmatic nucleus (SCN), encodes day length information by mechanisms that are not well understood. Here, we report that genetic ablation of miR-132/212 alters entrainment to different day lengths and non-24 hr day-night cycles, as well as photoperiodic regulation of Period2 expression in the SCN. SCN neurons from miR-132/212-deficient mice have significantly reduced dendritic spine density, along with altered methyl CpG-binding protein (MeCP2) rhythms. In Syrian hamsters, a model seasonal rodent, day length regulates spine density on SCN neurons in a melatonin-independent manner, as well as expression of miR-132, miR-212, and their direct target, MeCP2. Genetic disruption of Mecp2 fully restores the level of dendritic spines of miR-132/212-deficient SCN neurons. Our results reveal that, by regulating the dendritic structure of SCN neurons through a MeCP2-dependent mechanism, miR-132/212 affects the capacity of the SCN to encode seasonal time.


Assuntos
Adaptação Fisiológica/genética , Relógios Circadianos/genética , Dendritos/metabolismo , MicroRNAs/metabolismo , Estações do Ano , Adaptação Fisiológica/efeitos da radiação , Animais , Comportamento Animal , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Relógios Circadianos/efeitos da radiação , Dendritos/efeitos da radiação , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/efeitos da radiação , Feminino , Deleção de Genes , Regulação da Expressão Gênica/efeitos da radiação , Luz , Masculino , Mesocricetus , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Neurônios/metabolismo , Fotoperíodo , Proteoma/metabolismo , Transdução de Sinais/efeitos da radiação , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/efeitos da radiação , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo
7.
Proc Natl Acad Sci U S A ; 114(5): 1207-1212, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096421

RESUMO

Neuroendocrine mechanisms underlying social inhibition of puberty are not well understood. Here, we use a model exhibiting the most profound case of pubertal suppression among mammals to explore a role for RFamide-related peptide-3 [RFRP-3; mammalian ortholog to gonadotropin-inhibitory hormone (GnIH)] in neuroendocrine control of reproductive development. Naked mole rats (NMRs) live in sizable colonies where breeding is monopolized by two to four dominant animals, and no other members exhibit signs of puberty throughout their lives unless they are removed from the colony. Because of its inhibitory action on the reproductive axis in other vertebrates, we investigated the role of RFRP-3 in social reproductive suppression in NMRs. We report that RFRP-3 immunofluorescence expression patterns and RFRP-3/GnRH cross-talk are largely conserved in the NMR brain, with the exception of the unique presence of RFRP-3 cell bodies in the arcuate nucleus (Arc). Immunofluorescence comparisons revealed that central expression of RFRP-3 is altered by reproductive status, with RFRP-3 immunoreactivity enhanced in the paraventricular nucleus, dorsomedial nucleus, and Arc of reproductively quiescent NMRs. We further observed that exogenous RFRP-3 suppresses gonadal steroidogenesis and mating behavior in NMRs given the opportunity to undergo puberty. Together, our findings establish a role for RFRP-3 in preserving reproductive immaturity, and challenge the view that stimulatory peptides are the ultimate gatekeepers of puberty.


Assuntos
Sistema Límbico/metabolismo , Ratos-Toupeira/fisiologia , Neuropeptídeos/fisiologia , Maturidade Sexual/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Hipotalâmico Dorsomedial/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Injeções Intraventriculares , Kisspeptinas/metabolismo , Masculino , Neuropeptídeos/farmacologia , Ovário/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Progesterona/biossíntese , Progesterona/sangue , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia , Maturidade Sexual/efeitos dos fármacos , Isolamento Social , Testículo/metabolismo , Testosterona/biossíntese , Testosterona/sangue
8.
Cell Mol Life Sci ; 74(6): 1035-1059, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27689221

RESUMO

Circadian timekeeping is a ubiquitous mechanism that enables organisms to maintain temporal coordination between internal biological processes and time of the local environment. The molecular basis of circadian rhythms lies in a set of transcription-translation feedback loops (TTFLs) that drives the rhythmic transcription of core clock genes, whose level and phase of expression serve as the marker of circadian time. However, it has become increasingly evident that additional regulatory mechanisms impinge upon the TTFLs to govern the properties and behavior of the circadian clock. Such mechanisms include changes in chromatin architecture, interactions with other transcription factor networks, post-transcriptional control by RNA modifications, alternative splicing and microRNAs, and post-translational regulation of subcellular trafficking and protein degradation. In this review, we will summarize the current knowledge of circadian clock regulation-from transcriptional to post-translational-drawing from literature pertaining to the Drosophila and murine circadian systems.


Assuntos
Relógios Circadianos/genética , Drosophila melanogaster/genética , Animais , Regulação da Expressão Gênica , Camundongos , Modelos Genéticos , Biossíntese de Proteínas/genética , Transcrição Gênica
9.
Receptors Clin Investig ; 3(1)2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27088110

RESUMO

G protein-coupled receptor kinases (GRKs) are a family of serine/threonine protein kinases that terminate G protein-coupled receptor (GPCR) signaling by phosphorylating the receptor and inducing its internalization. In addition to their canonical function, some GRKs can phosphorylate non-GPCR substrates and regulate GPCR signaling in a kinase-independent manner. GPCRs are abundantly expressed in the suprachiasmatic nucleus (SCN), a structure in the mammalian brain that serves as the central circadian pacemaker. Various facets of circadian timekeeping are under the influence of GPCR signaling, and thus are potential targets for GRK regulation. Despite this, little attention has been given to the role of GRKs in circadian rhythms. In this research highlight, we discuss our latest findings on the functional involvement of GRK2 in mammalian circadian timekeeping in the SCN. Using grk2 knockout mice, we demonstrate that GRK2 is critical for maintaining proper clock speed and ensuring that the clock is appropriately synchronized to environmental light cycles. Although grk2 deficiency expectedly alters the expression of a key GPCR in the SCN, our study also reveals that GRK2 has a more direct function that touches the heart of the circadian clock.

10.
Cell Rep ; 12(8): 1272-88, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26279567

RESUMO

The pacemaker properties of the suprachiasmatic nucleus (SCN) circadian clock are shaped by mechanisms that influence the expression and behavior of clock proteins. Here, we reveal that G-protein-coupled receptor kinase 2 (GRK2) modulates the period, amplitude, and entrainment characteristics of the SCN. Grk2-deficient mice show phase-dependent alterations in light-induced entrainment, slower recovery from jetlag, and longer behavioral rhythms. Grk2 ablation perturbs intrinsic rhythmic properties of the SCN, increasing amplitude and decreasing period. At the cellular level, GRK2 suppresses the transcription of the mPeriod1 gene and the trafficking of PERIOD1 and PERIOD2 proteins to the nucleus. Moreover, GRK2 can physically interact with PERIOD1/2 and promote PERIOD2 phosphorylation at Ser545, effects that may underlie its ability to regulate PERIOD1/2 trafficking. Together, our findings identify GRK2 as an important modulator of circadian clock speed, amplitude, and entrainment by controlling PERIOD at the transcriptional and post-translational levels.


Assuntos
Núcleo Celular/metabolismo , Relógios Circadianos/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Proteínas Circadianas Period/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G/genética , Masculino , Camundongos , Proteínas Circadianas Period/genética , Fosforilação , Ligação Proteica
11.
J Neurosci ; 35(16): 6444-51, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904795

RESUMO

Polyglutamine (polyQ) expansion of the androgen receptor (AR) causes Kennedy's disease/spinobulbar muscular atrophy (KD/SBMA) through poorly defined cellular mechanisms. Although KD/SBMA has been thought of as a motor neuron disease, recent evidence indicates a key role for skeletal muscle. To resolve which early aspects of the disease can be caused by neurogenic or myogenic mechanisms, we made use of the tet-On and Cre-loxP genetic systems to selectively and acutely express polyQ AR in either motor neurons (NeuroAR) or myocytes (MyoAR) of transgenic mice. After 4 weeks of transgene induction in adulthood, deficits in gross motor function were seen in NeuroAR mice, but not MyoAR mice. Conversely, reduced size of fast glycolytic fibers and alterations in expression of candidate genes were observed only in MyoAR mice. Both NeuroAR and MyoAR mice exhibited reduced oxidative capacity in skeletal muscles, as well as a shift in fast fibers from oxidative to glycolytic. Markers of oxidative stress were increased in the muscle of NeuroAR mice and were reduced in motor neurons of both NeuroAR and MyoAR mice. Despite secondary pathology in skeletal muscle and behavioral deficits, no pathological signs were observed in motor neurons of NeuroAR mice, possibly due to relatively low levels of polyQ AR expression. These results indicate that polyQ AR in motor neurons can produce secondary pathology in muscle. Results also support both neurogenic and myogenic contributions of polyQ AR to several acute aspects of pathology and provide further evidence for disordered cellular respiration in KD/SBMA skeletal muscle.


Assuntos
Modelos Animais de Doenças , Neurônios Motores/patologia , Células Musculares/patologia , Transtornos Musculares Atróficos/patologia , Animais , Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Destreza Motora , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Transtornos Musculares Atróficos/genética , Estresse Oxidativo/genética , Receptores Androgênicos/genética
12.
Crit Rev Oncog ; 19(6): 505-16, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25597360

RESUMO

In 1984, a cytosolic protein was isolated from bovine brain and coined phosphatidylethanolamine binding protein (PEBP) to describe its phospholipid-binding potential. Its cellular function remained elusive for more than a decade until it was discovered that PEBP had the ability to suppress the Raf1-mitogen activated protein kinase (MAPK) pathway, earning it the new name of Raf1 kinase inhibitory protein (RKIP). This milestone discovery has paved the way for numerous studies that have now extended the reach of RKIP's function to other signaling cascades, within the context of various physiological and pathophysiological systems. This review will summarize our current knowledge of the neurophysiological roles of RKIP in the mammalian brain, including its function in the circadian clock and synaptic plasticity. It will also discuss evidence for an involvement of RKIP and its derived neuropeptide, hippocampal cholinergic neurostimulating peptide (HCNP), in neural development and differentiation. Implications in certain pathologies such as Alzheimer's disease and brain cancer will be highlighted. By chronicling the diverse functions of RKIP in the brain, we hope that this review will serve as a timely resource that ignites future studies on this versatile, multifaceted protein in the nervous system.


Assuntos
Encéfalo/metabolismo , Mamíferos , Proteína de Ligação a Fosfatidiletanolamina/fisiologia , Animais , Bovinos , Expressão Gênica , Humanos , Mamíferos/fisiologia , Proteína de Ligação a Fosfatidiletanolamina/química , Conformação Proteica , Distribuição Tecidual
13.
Cell Rep ; 5(4): 961-73, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24268780

RESUMO

The subgranular zone (SGZ) of the adult hippocampus contains a pool of quiescent neural progenitor cells (QNPs) that are capable of entering the cell cycle and producing newborn neurons. The mechanisms that control the timing and extent of adult neurogenesis are not well understood. Here, we show that QNPs of the adult SGZ express molecular-clock components and proliferate in a rhythmic fashion. The clock proteins PERIOD2 and BMAL1 are critical for proper control of neurogenesis. The absence of PERIOD2 abolishes the gating of cell-cycle entrance of QNPs, whereas genetic ablation of bmal1 results in constitutively high levels of proliferation and delayed cell-cycle exit. We use mathematical model simulations to show that these observations may arise from clock-driven expression of a cell-cycle inhibitor that targets the cyclin D/Cdk4-6 complex. Our findings may have broad implications for the circadian clock in timing cell-cycle events of other stem cell populations throughout the body.


Assuntos
Fatores de Transcrição ARNTL/genética , Relógios Circadianos/genética , Hipocampo/crescimento & desenvolvimento , Neurogênese/genética , Proteínas Circadianas Period/genética , Fatores de Transcrição ARNTL/biossíntese , Animais , Ciclo Celular/genética , Proliferação de Células , Ciclina D/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais , Neurônios/citologia , Proteínas Circadianas Period/biossíntese , Regiões Promotoras Genéticas
14.
Learn Mem ; 18(10): 610-6, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21921210

RESUMO

The immediate early gene (IEG) Arc is known to play an important role in synaptic plasticity; its protein is locally translated in the dendrites where it has been involved in several types of plasticity mechanisms. Because of its tight coupling with neuronal activity, Arc has been widely used as a tool to tag behaviorally activated networks. However, studies examining the modulation of Arc expression during and after learning have yielded somewhat contradictory results. Although some have reported that higher levels of Arc were induced by initial acquisition of a task rather than by reinstating a learned behavior, others have failed to observe such habituation of Arc transcription. Moreover, most of these studies have focused on the mRNA and, surprisingly, relatively little is known about how learning can affect Arc protein expression levels. Here we used taste recognition memory and examined Arc protein expression in the insular cortex of rats at distinct times during taste memory formation. Interestingly, we found that more Arc protein was induced by a familiar rather than by a novel taste. Moreover, this increase was inhibited by post-trial intrahippocampal anisomycin injections, a treatment known to inhibit safe-taste memory consolidation. In addition, confocal microscopy analysis of immunofluorescence stained tissue revealed that the proportion of IC neurons expressing Arc was the same in animals exposed to novel and familiar taste, but Arc immunoreactivity in dendrites was dramatically higher in rats exposed to the familiar taste. These results provide novel insights on how experience affects cortical plasticity.


Assuntos
Córtex Cerebral/citologia , Proteínas do Citoesqueleto/metabolismo , Dendritos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/ultraestrutura , Reconhecimento Psicológico/fisiologia , Paladar/fisiologia , Vias Aferentes/fisiologia , Animais , Anisomicina/farmacologia , Condicionamento Operante/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Masculino , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reconhecimento Psicológico/efeitos dos fármacos , Esquema de Reforço , Sacarina/administração & dosagem , Edulcorantes/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...