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










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(9): e0273098, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36107951

RESUMO

Naked mole-rats are a long-lived rodent species (current lifespan >37 years) and an increasingly popular biomedical model. Naked mole-rats exhibit neuroplasticity across their long lifespan. Previous studies have begun to investigate their neurogenic patterns. Here, we test the hypothesis that neuronal maturation is extended in this long-lived rodent. We characterize cell proliferation and neuronal maturation in established rodent neurogenic regions over 12 months following seven days of consecutive BrdU injection. Given that naked mole-rats are eusocial (high reproductive skew where only a few socially-dominant individuals reproduce), we also looked at proliferation in brain regions relevant to the social-decision making network. Finally, we measured co-expression of EdU (newly-born cells), DCX (immature neuron marker), and NeuN (mature neuron marker) to assess the timeline of neuronal maturation in adult naked mole-rats. This work reaffirms the subventricular zone as the main source of adult cell proliferation and suggests conservation of the rostral migratory stream in this species. Our profiling of socially-relevant brain regions suggests that future work which manipulates environmental context can unveil how newly-born cells integrate into circuitry and facilitate adult neuroplasticity. We also find naked mole-rat neuronal maturation sits at the intersection of rodents and long-lived, non-rodent species: while neurons can mature by 3 weeks (rodent-like), most neurons mature at 5 months and hippocampal neurogenic levels are low (like long-lived species). These data establish a timeline for future investigations of longevity- and socially-related manipulations of naked mole-rat adult neurogenesis.


Assuntos
Ratos-Toupeira , Neurogênese , Animais , Bromodesoxiuridina , Longevidade/fisiologia , Ratos-Toupeira/fisiologia , Neurônios/fisiologia
2.
Nat Commun ; 13(1): 1594, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35332162

RESUMO

Ubiquitin ligases control the degradation of core clock proteins to govern the speed and resetting properties of the circadian pacemaker. However, few studies have addressed their potential to regulate other cellular events within clock neurons beyond clock protein turnover. Here, we report that the ubiquitin ligase, UBR4/POE, strengthens the central pacemaker by facilitating neuropeptide trafficking in clock neurons and promoting network synchrony. Ubr4-deficient mice are resistant to jetlag, whereas poe knockdown flies are prone to arrhythmicity, behaviors reflective of the reduced axonal trafficking of circadian neuropeptides. At the cellular level, Ubr4 ablation impairs the export of secreted proteins from the Golgi apparatus by reducing the expression of Coronin 7, which is required for budding of Golgi-derived transport vesicles. In summary, UBR4/POE fulfills a conserved and unexpected role in the vesicular trafficking of neuropeptides, a function that has important implications for circadian clock synchrony and circuit-level signal processing.


Assuntos
Relógios Circadianos , Proteínas de Drosophila , Neuropeptídeos , Animais , Proteínas CLOCK/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Relógios Circadianos/genética , Ritmo Circadiano , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Camundongos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
3.
Neurol Int ; 13(4): 541-554, 2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34842772

RESUMO

Mood disorders negatively impact the lives of hundreds of millions of individuals worldwide every year, yet the precise molecular mechanisms by which they manifest remain elusive. Circadian dysregulation is one avenue by which mood disorders are thought to arise. SOX2 is a transcription factor that is highly expressed in the murine suprachiasmatic nucleus (SCN), the circadian master clock, and has been recently found to be an important regulator of Per2, a core component of the molecular clock. Genetic ablation of the Sox2 gene in GABAergic neurons selectively impacts SCN neurons, as they are one of very few, if not the only, GABAergic populations that express Sox2. Here, we show that GABAergic-restricted ablation of Sox2 results in anxio-depressive-like phenotypes in mice as observed in the elevated plus maze, forced swim test, tail suspension test, and sucrose preference test. We further observe a reduction in basal and/or forced swim-induced c-Fos expression, a marker of neuronal activation, in the nucleus incertus, arcuate nucleus, and dentate gyrus of Sox2 conditional knockout (cKO) mice. Given the restricted disruption of SOX2 expression in the SCN of Sox2 cKO mice, we propose that their mood-associated phenotypes are the consequence of a dysregulated central clock that is unable to communicate appropriately timed signals to other brain nuclei that regulate affective behaviors.

4.
Cell Rep ; 36(11): 109704, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525369

RESUMO

Histone variants are crucial regulators of chromatin structure and gene transcription, yet their functions within the brain remain largely unexplored. Here, we show that the H2A histone variant H2A.Z is essential for neuronal survival. Mice lacking H2A.Z in GABAergic neurons or Purkinje cells (PCs) present with a progressive cerebellar ataxia accompanied by widespread degeneration of PCs. Ablation of H2A.Z in other neuronal subtypes also triggers cell death. H2A.Z binds to the promoters of key nuclear-encoded mitochondrial genes to regulate their expression and promote organelle function. Bolstering mitochondrial activity genetically or by organelle transplant enhances the survival of H2A.Z-ablated neurons. Changes in bioenergetic status alter H2A.Z occupancy at the promoters of nuclear-encoded mitochondrial genes, an adaptive response essential for cell survival. Our results highlight that H2A.Z fulfills a key, conserved role in neuronal survival by acting as a transcriptional rheostat to regulate the expression of genes critical to mitochondrial function.


Assuntos
Núcleo Celular/metabolismo , Histonas/genética , Mitocôndrias/metabolismo , Transcriptoma , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Fibroblastos/citologia , Fibroblastos/metabolismo , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Histonas/deficiência , Histonas/metabolismo , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fosforilação Oxidativa , Células de Purkinje/citologia , Células de Purkinje/metabolismo , Transcriptoma/efeitos dos fármacos , Regulação para Cima
5.
Front Neurosci ; 15: 659974, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33833665

RESUMO

The suprachiasmatic nucleus (SCN) of the hypothalamus is the central circadian clock of mammals. It is responsible for communicating temporal information to peripheral oscillators via humoral and endocrine signaling, ultimately controlling overt rhythms such as sleep-wake cycles, body temperature, and locomotor activity. Given the heterogeneity and complexity of the SCN, its genesis is tightly regulated by countless intrinsic and extrinsic factors. Here, we provide a brief overview of the development of the SCN, with special emphasis on the murine system.

6.
Int J Mol Sci ; 23(1)2021 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-35008655

RESUMO

In mammals, the hypothalamic suprachiasmatic nucleus (SCN) functions as the central circadian pacemaker, orchestrating behavioral and physiological rhythms in alignment to the environmental light/dark cycle. The neurons that comprise the SCN are anatomically and functionally heterogeneous, but despite their physiological importance, little is known about the pathways that guide their specification and differentiation. Here, we report that the stem/progenitor cell transcription factor, Sex determining region Y-box 2 (Sox2), is required in the embryonic SCN to control the expression of SCN-enriched neuropeptides and transcription factors. Ablation of Sox2 in the developing SCN leads to downregulation of circadian neuropeptides as early as embryonic day (E) 15.5, followed by a decrease in the expression of two transcription factors involved in SCN development, Lhx1 and Six6, in neonates. Thymidine analog-retention assays revealed that Sox2 deficiency contributed to reduced survival of SCN neurons during the postnatal period of cell clearance, but did not affect progenitor cell proliferation or SCN specification. Our results identify SOX2 as an essential transcription factor for the proper differentiation and survival of neurons within the developing SCN.


Assuntos
Diferenciação Celular , Desenvolvimento Embrionário , Neurônios/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano , Camundongos , Neurônios/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Núcleo Supraquiasmático/crescimento & desenvolvimento , Núcleo Supraquiasmático/fisiologia
7.
J Biol Rhythms ; 34(6): 634-644, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31452438

RESUMO

The principal circadian pacemaker in mammals, the suprachiasmatic nucleus (SCN), expresses a number of neuropeptides that facilitate intercellular synchrony, helping to generate coherent outputs to peripheral clocks throughout the body. In particular, arginine vasopressin (AVP)- and vasoactive intestinal peptide (VIP)-expressing neurons have been recognized as crucial subpopulations within the SCN and have thus been the focus of many chronobiological studies. Here, we analyze the neuropeptide expression of 2 popular transgenic mouse strains commonly used to direct or restrict Cre-mediated recombination to AVP- and VIP-ergic neurons. The Avp-IRES2-Cre (JAX #023530) and Vip-IRES-Cre (JAX #010908) "driver" mouse strains express the Cre recombinase under the control of the endogenous Avp or Vip gene, respectively, allowing scientists either to ablate their gene of interest or to overexpress a transgene in a cell type-specific manner. Although these are potentially very powerful tools for chronobiologists and other scientists studying AVP- and VIP-ergic neurons, we found that neuropeptide expression in these mice is significantly decreased when an IRES(2)-Cre cassette is inserted downstream of the neuropeptide-encoding gene locus. The impact of IRES(2)-Cre cassette insertion on neuropeptide expression may be a confounding factor in many experimental designs. Our findings suggest that extreme caution must be exercised when using these mouse models to avoid misinterpretation of empirical results.


Assuntos
Arginina Vasopressina/genética , Relógios Circadianos , Expressão Gênica , Camundongos Transgênicos , Peptídeo Intestinal Vasoativo/genética , Animais , Fenômenos Cronobiológicos , Ritmo Circadiano , Feminino , Integrases/genética , Masculino , Camundongos , Neurônios/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia
8.
Data Brief ; 24: 103909, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31061857

RESUMO

SOX2 is a stem cell-associated pluripotency transcription factor whose role in neuronal populations is undefined. Here we present the RNA-sequencing based transcriptome profiles of control (Sox2 fl/fl ) and SOX2 conditional knock-out (Vgat-cre;Sox2 fl/fl ) mice at four time points in one 24-h circadian cycle. The raw sequencing data were deposited to ArrayExpress database at EMBL-EBI (https://www.ebi.ac.uk/arrayexpress) under the accession number E-MTAB-7496. Results of rhythmicity analysis, differential expression analysis, network prediction, and potential target identification stemming from the RNA-sequencing dataset are also given in this article. The interpretation and discussion of these data can be found in the related research article entitled "SOX2-dependent transcription in clock neurons promotes the robustness of the central circadian pacemaker." Cheng et al. 2019.

9.
Int J Mol Sci ; 20(9)2019 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-31086044

RESUMO

The central pacemakers of circadian timekeeping systems are highly robust yet adaptable, providing the temporal coordination of rhythms in behavior and physiological processes in accordance with the demands imposed by environmental cycles. These features of the central pacemaker are achieved by a multi-oscillator network in which individual cellular oscillators are tightly coupled to the environmental day-night cycle, and to one another via intercellular coupling. In this review, we will summarize the roles of various neurotransmitters and neuropeptides in the regulation of circadian entrainment and synchrony within the mammalian and Drosophila central pacemakers. We will also describe the diverse functions of protein kinases in the relay of input signals to the core oscillator or the direct regulation of the molecular clock machinery.


Assuntos
Ritmo Circadiano/fisiologia , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Drosophila , Humanos , Camundongos , Núcleo Supraquiasmático/metabolismo
10.
Cell Rep ; 26(12): 3191-3202.e8, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893593

RESUMO

Clock neurons within the mammalian suprachiasmatic nuclei (SCN) encode circadian time using interlocked transcription-translation feedback loops (TTFLs) that drive rhythmic gene expression. However, the contributions of other transcription factors outside of the circadian TTFLs to the functionality of the SCN remain obscure. Here, we report that the stem and progenitor cell transcription factor, sex-determining region Y-box 2 (SOX2), is expressed in adult SCN neurons and positively regulates transcription of the core clock gene, Period2. Mice lacking SOX2 selectively in SCN neurons display imprecise, poorly consolidated behavioral rhythms that do not entrain efficiently to environmental light cycles and that are highly susceptible to constant light-induced arrhythmicity. RNA sequencing revealed that Sox2 deficiency alters the SCN transcriptome, reducing the expression of core clock genes and neuropeptide-receptor systems. By defining the transcriptional landscape within SCN neurons, SOX2 enables the generation of robust, entrainable circadian rhythms that accurately reflect environmental time.


Assuntos
Relógios Circadianos/fisiologia , Proteínas Circadianas Period/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Núcleo Supraquiasmático/metabolismo , Transcrição Gênica , Animais , Camundongos , Camundongos Transgênicos , Proteínas Circadianas Period/genética , Fatores de Transcrição SOXB1/genética , Núcleo Supraquiasmático/citologia
11.
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
12.
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
13.
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.

14.
Int J Dev Biol ; 59(7-9): 313-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26679948

RESUMO

We have recently characterized essential inositol 1,4,5-trisphosphate receptor (IP 3R) and ryanodine receptor (RyR)-mediated Ca(2+) signals generated during the differentiation of slow muscle cells (SMCs) in intact zebrafish embryos. Here, we show that the lysosomal two-pore channel 2 (TPC2) also plays a crucial role in generating, and perhaps triggering, these essential Ca(2+) signals, and thus contributes to the regulation of skeletal muscle myogenesis. We used a transgenic line of zebrafish that expresses the bioluminescent Ca(2+) reporter, aequorin, specifically in skeletal muscle, in conjunction with morpholino (MO)-based and pharmacological inhibition of TPC2, in both intact embryos and isolated SMCs. MO-based knock-down of TPC2 resulted in a dramatic attenuation of the Ca(2+) signals, whereas the introduction of TPCN2-MO and TPCN2 mRNA together partially rescued the Ca(2+) signaling signature. Embryos treated with trans-ned-19 or bafilomycin A1, a specific NAADP receptor inhibitor and vacuolar-type H(+)ATPase inhibitor, respectively, also displayed a similar disruption of SMC Ca(2+) signaling. TPC2 and lysosomes were shown via immunohistochemistry and confocal laser scanning microscopy to be localized in perinuclear and striated cytoplasmic domains of SMCs, coincident with patterns of IP 3R and RyR expression. These data together imply that TPC2-mediated Ca(2+) release from lysosomes acts upstream from RyR- and IP 3R-mediated Ca(2+) release, suggesting that the former might act as a sensitive trigger to initiate the SR-mediated Ca(2+)-induced-Ca(2+)-release essential for SMC myogenesis and function.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/genética , Cálcio/metabolismo , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Canais de Cálcio/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Lisossomos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
15.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...