Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Genet ; 15: 1343030, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38818037

RESUMEN

Circadian rhythms, essential 24-hour cycles guiding biological functions, synchronize organisms with daily environmental changes. These rhythms, which are evolutionarily conserved, govern key processes like feeding, sleep, metabolism, body temperature, and endocrine secretion. The central clock, located in the suprachiasmatic nucleus (SCN), orchestrates a hierarchical network, synchronizing subsidiary peripheral clocks. At the cellular level, circadian expression involves transcription factors and epigenetic remodelers, with environmental signals contributing flexibility. Circadian disruption links to diverse diseases, emphasizing the urgency to comprehend the underlying mechanisms. This review explores the communication between the environment and chromatin, focusing on histone post-translational modifications. Special attention is given to the significance of histone methylation in circadian rhythms and metabolic control, highlighting its potential role as a crucial link between metabolism and circadian rhythms. Understanding these molecular intricacies holds promise for preventing and treating complex diseases associated with circadian disruption.

2.
Mol Neurobiol ; 2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38340204

RESUMEN

Stem cells possess extraordinary capacities for self-renewal and differentiation, making them highly valuable in regenerative medicine. Among these, neural stem cells (NSCs) play a fundamental role in neural development and repair processes. NSC characteristics and fate are intricately regulated by the microenvironment and intracellular signaling. Interestingly, metabolism plays a pivotal role in orchestrating the epigenome dynamics during neural differentiation, facilitating the transition from undifferentiated NSC to specialized neuronal and glial cell types. This intricate interplay between metabolism and the epigenome is essential for precisely regulating gene expression patterns and ensuring proper neural development. This review highlights the mechanisms behind metabolic regulation of NSC fate and their connections with epigenetic regulation to shape transcriptional programs of stemness and neural differentiation. A comprehensive understanding of these molecular gears appears fundamental for translational applications in regenerative medicine and personalized therapies for neurological conditions.

3.
Nat Commun ; 14(1): 1685, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36973248

RESUMEN

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.


Asunto(s)
Relojes Circadianos , Enfermedades Metabólicas , Ratones , Masculino , Femenino , Animales , Ritmo Circadiano/fisiología , NAD/metabolismo , Dieta , Enfermedades Metabólicas/metabolismo , Hígado/metabolismo
4.
Glia ; 71(7): 1626-1647, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36919670

RESUMEN

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.


Asunto(s)
Relojes Circadianos , Ratones , Animales , Relojes Circadianos/genética , Astrocitos/metabolismo , Hipotálamo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Metabolismo Energético/genética
5.
Nat Cardiovasc Res ; 2(2): 174-191, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38665902

RESUMEN

Cardiac metabolism is deranged in heart failure, but underlying mechanisms remain unclear. Here, we show that lysine demethylase 8 (Kdm8) maintains an active mitochondrial gene network by repressing Tbx15, thus preventing dilated cardiomyopathy leading to lethal heart failure. Deletion of Kdm8 in mouse cardiomyocytes increased H3K36me2 with activation of Tbx15 and repression of target genes in the NAD+ pathway before dilated cardiomyopathy initiated. NAD+ supplementation prevented dilated cardiomyopathy in Kdm8 mutant mice, and TBX15 overexpression blunted NAD+-activated cardiomyocyte respiration. Furthermore, KDM8 was downregulated in human hearts affected by dilated cardiomyopathy, and higher TBX15 expression defines a subgroup of affected hearts with the strongest downregulation of genes encoding mitochondrial proteins. Thus, KDM8 represses TBX15 to maintain cardiac metabolism. Our results suggest that epigenetic dysregulation of metabolic gene networks initiates myocardium deterioration toward heart failure and could underlie heterogeneity of dilated cardiomyopathy.

6.
Cells ; 11(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36359853

RESUMEN

Organotypic three-dimensional (3D) cell cultures more accurately mimic the characteristics of solid tumors in vivo in comparison with traditional two-dimensional (2D) monolayer cell models. Currently, studies on the regulation of long non-coding RNAs (lncRNAs) have not been explored in breast cancer cells cultured in 3D microenvironments. In the present research, we studied the expression and potential roles of lncRNAs in estrogen receptor-positive luminal B subtype BT-474 breast cancer cells grown over extracellular matrix proteins-enriched 3D cultures. Global expression profiling using DNA microarrays identifies 290 upregulated and 183 downregulated lncRNAs in 3D cultures relative to 2D condition. Using a co-expression analysis approach of lncRNAs and mRNAs pairs expressed in the same experimental conditions, we identify hundreds of regulatory axes modulating genes involved in cancer hallmarks, such as responses to estrogens, cell proliferation, hypoxia, apical junctions, and resistance to endocrine therapy. In addition, we identified 102 lncRNAs/mRNA correlations in 3D cultures, which were similar to those reported in TCGA datasets obtained from luminal B breast cancer patients. Interestingly, we also found a set of mRNAs transcripts co-expressed with LINC00847 and CTD-2566J3.1 lncRNAs, which were predictors of pathologic complete response and overall survival. Finally, both LINC00847 and CTD -2566J3.1 were co-expressed with essential genes for cancer genetic dependencies, such as FOXA1 y GINS2. Our experimental and predictive findings show that co-expressed lncRNAs/mRNAs pairs exhibit a high degree of similarity with those found in luminal B breast cancer patients, suggesting that they could be adequate pre-clinical tools to identify not only biomarkers related to endocrine therapy response and PCR, but to understand the biological behavior of cancer cells in 3D microenvironments.


Asunto(s)
Neoplasias de la Mama , ARN Largo no Codificante , Humanos , Femenino , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Oncogenes , Carcinogénesis/genética , Microambiente Tumoral/genética , Proteínas Cromosómicas no Histona/metabolismo
7.
J Cell Biol ; 221(12)2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36197339

RESUMEN

Adipocytes are the main cell type in adipose tissue, which is a critical regulator of metabolism, highly specialized in storing energy as fat. Adipocytes differentiate from multipotent mesenchymal stromal cells (hMSCs) through adipogenesis, a tightly controlled differentiation process involving close interplay between metabolic transitions and sequential programs of gene expression. However, the specific gears driving this interplay remain largely obscure. Additionally, the metabolite nicotinamide adenine dinucleotide (NAD+) is becoming increasingly recognized as a regulator of lipid metabolism, and a promising therapeutic target for dyslipidemia and obesity. Here, we explored how NAD+ bioavailability controls adipogenic differentiation from hMSC. We found a previously unappreciated repressive role for NAD+ on adipocyte commitment, while a functional NAD+-dependent deacetylase SIRT1 appeared crucial for terminal differentiation of pre-adipocytes. Repressing NAD+ biosynthesis during adipogenesis promoted the adipogenic transcriptional program, while two-photon microscopy and extracellular flux analyses suggest that SIRT1 activity mostly relies on the metabolic switch. Interestingly, SIRT1 controls subcellular compartmentalization of redox metabolism during adipogenesis.


Asunto(s)
Adipocitos , Adipogénesis , NAD , Sirtuina 1 , Adipocitos/metabolismo , Diferenciación Celular , Expresión Génica , NAD/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo
8.
Cells ; 11(1)2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-35011637

RESUMEN

A growing body of research on the transcriptome and cancer genome has demonstrated that many gynecological tumor-specific gene mutations are located in cis-regulatory elements. Through chromosomal looping, cis-regulatory elements interact which each other to control gene expression by bringing distant regulatory elements, such as enhancers and insulators, into close proximity with promoters. It is well known that chromatin connections may be disrupted in cancer cells, promoting transcriptional dysregulation and the expression of abnormal tumor suppressor genes and oncogenes. In this review, we examine the roles of alterations in 3D chromatin interactions. This includes changes in CTCF protein function, cancer-risk single nucleotide polymorphisms, viral integration, and hormonal response as part of the mechanisms that lead to the acquisition of enhancers or super-enhancers. The translocation of existing enhancers, as well as enhancer loss or acquisition of insulator elements that interact with gene promoters, is also revised. Remarkably, similar processes that modify 3D chromatin contacts in gene promoters may also influence the expression of non-coding RNAs, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), which have emerged as key regulators of gene expression in a variety of cancers, including gynecological malignancies.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Cromatina/metabolismo , Neoplasias de los Genitales Femeninos/genética , Genoma Humano , Transcripción Genética , Animales , Neoplasias de la Mama/patología , Carcinogénesis/patología , Femenino , Neoplasias de los Genitales Femeninos/patología , Humanos
9.
Artículo en Inglés | MEDLINE | ID: mdl-32232012

RESUMEN

The circadian clock orchestrates daily rhythms in many physiological, behavioral and molecular processes, providing means to anticipate, and adapt to environmental changes. A specific role of the circadian clock is to coordinate functions of the immune system both at steady-state and in response to infectious threats. Hence, time-of-day dependent variables are found in the physiology of immune cells, host-parasite interactions, inflammatory processes, or adaptive immune responses. Interestingly, the molecular clock coordinates transcriptional-translational feedback loops which orchestrate daily oscillations in expression of many genes involved in cellular functions. This clock function is assisted by tightly controlled transitions in the chromatin fiber involving epigenetic mechanisms which determine how a when transcriptional oscillations occur. Immune cells are no exception, as they also present a functional clock dictating transcriptional rhythms. Hereby, the molecular clock and the chromatin regulators controlling rhythmicity represent a unique scaffold mediating the crosstalk between the circadian and the immune systems. Certain epigenetic regulators are shared between both systems and uncovering them and characterizing their dynamics can provide clues to design effective chronotherapeutic strategies for modulation of the immune system.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Cromatina , Relojes Circadianos/genética , Epigénesis Genética , Epigenómica
10.
Clin Epigenetics ; 11(1): 79, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092281

RESUMEN

Circadian rhythms orchestrate crucial physiological functions and behavioral aspects around a day in almost all living forms. The circadian clock is a time tracking system that permits organisms to predict and anticipate periodic environmental fluctuations. The circadian system is hierarchically organized, and a master pacemaker located in the brain synchronizes subsidiary clocks in the rest of the organism. Adequate synchrony between central and peripheral clocks ensures fitness and potentiates a healthy state. Conversely, disruption of circadian rhythmicity is associated with metabolic diseases, psychiatric disorders, or cancer, amongst other pathologies. Remarkably, the molecular machinery directing circadian rhythms consists of an intricate network of feedback loops in transcription and translation which impose 24-h cycles in gene expression across all tissues. Interestingly, the molecular clock collaborates with multitude of epigenetic remodelers to fine tune transcriptional rhythms in a tissue-specific manner. Very exciting research demonstrate that three-dimensional properties of the genome have a regulatory role on circadian transcriptional rhythmicity, from bacteria to mammals. Unexpectedly, highly dynamic long-range chromatin interactions have been revealed during the circadian cycle in mammalian cells, where thousands of regulatory elements physically interact with promoter regions every 24 h. Molecular mechanisms directing circadian dynamics on chromatin folding are emerging, and the coordinated action between the core clock and epigenetic remodelers appears to be essential for these movements. These evidences reveal a critical epigenetic regulatory layer for circadian rhythms and pave the way to uncover molecular mechanisms triggering pathological states associated to circadian misalignment.


Asunto(s)
Cromatina/química , Ritmo Circadiano , Transcripción Genética , Animales , Cromatina/genética , Relojes Circadianos , Epigénesis Genética , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Aptitud Genética , Humanos , Especificidad de Órganos
11.
Biol Psychiatry ; 82(5): 351-360, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28395871

RESUMEN

BACKGROUND: Conventional antidepressants usually require several weeks to achieve a full clinical response in patients with major depressive disorder, an illness associated with dysregulated circadian rhythms and a high incidence of suicidality. Two rapid-acting antidepressant strategies, low-dose ketamine (KT) and sleep deprivation (SD) therapies, dramatically reduce depressive symptoms within 24 hours in a subset of major depressive disorder patients. However, it is unknown whether they exert their actions through shared regulatory mechanisms. To address this question, we performed comparative transcriptomics analyses to identify candidate genes and relevant pathways common to KT and SD. METHODS: We used the forced swim test, a standardized behavioral approach to measure antidepressant-like activity of KT and SD. We investigated gene expression changes using high-density microarrays and pathway analyses (Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, Gene Set Enrichment Analysis) in KT- and SD-treated mice compared with saline-treated control male mice. RESULTS: We show that KT and SD elicit common transcriptional responses implicating distinct elements of the circadian clock and processes involved in neuronal plasticity. There is an overlap of 64 genes whose expression is common in KT and SD. Specifically, there is downregulation of clock genes including Ciart, Per2, Npas4, Dbp, and Rorb in both KT- and SD-treated mice. CONCLUSIONS: We demonstrate a potential involvement of the circadian clock in rapid antidepressant responses. These findings could open new research avenues to help design chronopharmacological strategies to treat major depressive disorder.


Asunto(s)
Antidepresivos/farmacología , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Trastorno Depresivo/terapia , Giro del Cíngulo/metabolismo , Ketamina/farmacología , Privación de Sueño/metabolismo , Animales , Biología Computacional , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Giro del Cíngulo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Análisis por Micromatrices , Transcriptoma/efectos de los fármacos , Transcriptoma/fisiología
12.
Proc Natl Acad Sci U S A ; 113(45): 12715-12720, 2016 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-27791113

RESUMEN

Sirtuin 1 (SIRT1) is an NAD+-dependent deacetylase that functions as metabolic sensor of cellular energy and modulates biochemical pathways in the adaptation to changes in the environment. SIRT1 substrates include histones and proteins related to enhancement of mitochondrial function as well as antioxidant protection. Fluctuations in intracellular NAD+ levels regulate SIRT1 activity, but how SIRT1 enzymatic activity impacts on NAD+ levels and its intracellular distribution remains unclear. Here, we show that SIRT1 determines the nuclear organization of protein-bound NADH. Using multiphoton microscopy in live cells, we show that free and bound NADH are compartmentalized inside of the nucleus, and its subnuclear distribution depends on SIRT1. Importantly, SIRT6, a chromatin-bound deacetylase of the same class, does not influence NADH nuclear localization. In addition, using fluorescence fluctuation spectroscopy in single living cells, we reveal that NAD+ metabolism in the nucleus is linked to subnuclear dynamics of active SIRT1. These results reveal a connection between NAD+ metabolism, NADH distribution, and SIRT1 activity in the nucleus of live cells and pave the way to decipher links between nuclear organization and metabolism.

13.
Biochim Biophys Acta ; 1859(10): 1314-21, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27345571

RESUMEN

It is well established that eukaryotic genomes are pervasively transcribed producing cryptic unstable transcripts (CUTs). However, the mechanisms regulating pervasive transcription are not well understood. Here, we report that the fission yeast CENP-B homolog Abp1 plays an important role in preventing pervasive transcription. We show that loss of abp1 results in the accumulation of CUTs, which are targeted for degradation by the exosome pathway. These CUTs originate from different types of genomic features, but the highest increase corresponds to Tf2 retrotransposons and rDNA repeats, where they map along the entire elements. In the absence of abp1, increased RNAPII-Ser5P occupancy is observed throughout the Tf2 coding region and, unexpectedly, RNAPII-Ser5P is enriched at rDNA repeats. Loss of abp1 also results in Tf2 derepression and increased nucleolus size. Altogether these results suggest that Abp1 prevents pervasive RNAPII transcription of repetitive DNA elements (i.e., Tf2 and rDNA repeats) from internal cryptic sites.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación Fúngica de la Expresión Génica , ARN Polimerasa II/genética , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Transcripción Genética , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestructura , Centrómero/metabolismo , Centrómero/ultraestructura , Proteína B del Centrómero/genética , Proteína B del Centrómero/metabolismo , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Proteínas de Unión al ADN/deficiencia , Heterocromatina/metabolismo , Heterocromatina/ultraestructura , ARN Polimerasa II/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Retroelementos , Schizosaccharomyces/metabolismo , Schizosaccharomyces/ultraestructura , Proteínas de Schizosaccharomyces pombe/metabolismo
14.
Cell Metab ; 23(3): 467-78, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26959185

RESUMEN

Organismal homeostasis relies on coherent interactions among tissues, specifically between brain-driven functions and peripheral metabolic organs. Hypothalamic circuits compute metabolic information to optimize energetic resources, but the role of the circadian clock in these pathways remains unclear. We have generated mice with targeted ablation of the core-clock gene Bmal1 within Sf1-neurons of the ventromedial hypothalamus (VMH). While this mutation does not affect the central clock in the suprachiasmatic nucleus (SCN), the VMH clock controls cyclic thermogenesis in brown adipose tissue (BAT), a tissue that governs energy balance by dissipating chemical energy as heat. VMH-driven control is exerted through increased adrenergic signaling within the sympathetic nervous system, without affecting the BAT's endogenous clock. Moreover, we show that the VMH circadian clock computes light and feeding inputs to modulate basal energy expenditure. Thus, we reveal a previously unsuspected circuit where an SCN-independent, hypothalamic circadian clock controls BAT function, energy expenditure, and thermogenesis.


Asunto(s)
Metabolismo Energético , Núcleo Supraquiasmático/fisiología , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Relojes Circadianos , Ritmo Circadiano , Homeostasis , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Proteínas Circadianas Period/fisiología , Sistema Nervioso Simpático/fisiología , Termogénesis
15.
Nat Struct Mol Biol ; 22(4): 312-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25751424

RESUMEN

The circadian clock controls the transcription of hundreds of genes through specific chromatin-remodeling events. The histone methyltransferase mixed-lineage leukemia 1 (MLL1) coordinates recruitment of CLOCK-BMAL1 activator complexes to chromatin, an event associated with cyclic trimethylation of histone H3 Lys4 (H3K4) at circadian promoters. Remarkably, in mouse liver circadian H3K4 trimethylation is modulated by SIRT1, an NAD(+)-dependent deacetylase involved in clock control. We show that mammalian MLL1 is acetylated at two conserved residues, K1130 and K1133. Notably, MLL1 acetylation is cyclic, controlled by the clock and by SIRT1, and it affects the methyltransferase activity of MLL1. Moreover, H3K4 methylation at clock-controlled-gene promoters is influenced by pharmacological or genetic inactivation of SIRT1. Finally, levels of MLL1 acetylation and H3K4 trimethylation at circadian gene promoters depend on NAD(+) circadian levels. These findings reveal a previously unappreciated regulatory pathway between energy metabolism and histone methylation.


Asunto(s)
Relojes Circadianos/genética , Histonas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , NAD/fisiología , Sirtuina 1/fisiología , Acetilación , Animales , Cromatina , Regulación de la Expresión Génica , Metilación , Ratones , Modelos Genéticos , NAD/metabolismo , Sirtuina 1/metabolismo
16.
Curr Mol Biol Rep ; 1(1): 1-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27014564

RESUMEN

The molecular circadian clock orchestrates the daily cyclical expression of thousands of genes. Disruption of this transcriptional program leads to a variety of pathologies, including insomnia, depression and metabolic disorders. Circadian rhythms in gene expression rely on specific chromatin transitions which are ultimately coordinated by the molecular clock. As a consequence, a highly plastic and dynamic circadian epigenome can be delineated across different tissues and cell types. Intriguingly, genome topology appears to coordinate cyclic transcription at circadian interactomes, in which circadian genes are in physical contact within the cell nucleus in a time-specific manner. Moreover, the clock machinery shows functional interplays with key metabolic regulators, thereby connecting the circadian epigenome to cellular metabolism. Unraveling the molecular aspects of such interplays is likely to reveal new therapeutic strategies towards the treatment of metabolic disorders.

17.
Proc Natl Acad Sci U S A ; 112(22): 6863-70, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25378702

RESUMEN

Circadian rhythms drive the temporal organization of a wide variety of physiological and behavioral functions in ∼24-h cycles. This control is achieved through a complex program of gene expression. In mammals, the molecular clock machinery consists of interconnected transcriptional-translational feedback loops that ultimately ensure the proper oscillation of thousands of genes in a tissue-specific manner. To achieve circadian transcriptional control, chromatin remodelers serve the clock machinery by providing appropriate oscillations to the epigenome. Recent findings have revealed the presence of circadian interactomes, nuclear "hubs" of genome topology where coordinately expressed circadian genes physically interact in a spatial and temporal-specific manner. Thus, a circadian nuclear landscape seems to exist, whose interplay with metabolic pathways and clock regulators translates into specific transcriptional programs. Deciphering the molecular mechanisms that connect the circadian clock machinery with the nuclear landscape will reveal yet unexplored pathways that link cellular metabolism to epigenetic control.


Asunto(s)
Cromatina/metabolismo , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Epigénesis Genética/fisiología , Mamíferos/fisiología , Modelos Biológicos , Transcripción Genética/fisiología , Animales , Relojes Circadianos/genética , Retroalimentación Fisiológica/fisiología , Redes y Vías Metabólicas/fisiología , Sirtuinas/metabolismo
18.
Nat Struct Mol Biol ; 20(10): 1206-13, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24056944

RESUMEN

Dynamic transitions in the epigenome have been associated with regulated patterns of nuclear organization. The accumulating evidence that chromatin remodeling is implicated in circadian function prompted us to explore whether the clock may control nuclear architecture. We applied the chromosome conformation capture on chip technology in mouse embryonic fibroblasts (MEFs) to demonstrate the presence of circadian long-range interactions using the clock-controlled Dbp gene as bait. The circadian genomic interactions with Dbp were highly specific and were absent in MEFs whose clock was disrupted by ablation of the Bmal1 gene (also called Arntl). We establish that the Dbp circadian interactome contains a wide variety of genes and clock-related DNA elements. These findings reveal a previously unappreciated circadian and clock-dependent shaping of the nuclear landscape.


Asunto(s)
Cromosomas , Relojes Circadianos , Factores de Transcripción ARNTL/fisiología , Animales , Células Cultivadas , Ratones
19.
Curr Opin Cell Biol ; 25(2): 170-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23385084

RESUMEN

Circadian rhythms occur in most of the living organisms, and with a 24 hour periodicity govern a number of physiological and metabolic functions. During the past few years, an important research effort has uncovered new trails that intersect between circadian rhythms and metabolic pathways. At a molecular level, the clock machinery is responsible for the establishment of a circadian epigenome, and this can be modulated by metabolic cues. Indeed, metabolic control by the circadian clock is manifest in the development of metabolic diseases when circadian rhythms are impaired. Thus, pharmacological modulation of circadian rhythms promises new avenues for the treatment of metabolic and sleep disorders.


Asunto(s)
Ensamble y Desensamble de Cromatina , Relojes Circadianos/genética , Ritmo Circadiano/genética , Metabolismo Energético/genética , Epigénesis Genética , Animales , Ensamble y Desensamble de Cromatina/genética , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Epigenómica , Humanos , Enfermedades Metabólicas/genética , Trastornos del Sueño-Vigilia/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA