Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38854114

RESUMEN

The circadian clock orchestrates vital physiological processes such as metabolism, immune function, and tissue regeneration, aligning them with the optimal time of day. This study identifies an intricate interplay between the circadian clock within muscle stem cells (SCs) and their capacity to modulate the immune microenvironment during muscle regeneration. We uncover that the SC clock provokes time of day-dependent induction of inflammatory response genes following injury, particularly those related to neutrophil activity and chemotaxis. These responses are driven by rhythms of cytosolic regeneration of the signaling metabolite NAD+. We demonstrate that genetically enhancing cytosolic NAD+ regeneration in SCs is sufficient to induce robust inflammatory responses that significantly influence muscle regeneration. Furthermore, using mononuclear single-cell sequencing of the regenerating muscle niche, we uncover a key role for the cytokine CCL2 in mediating SC-neutrophil crosstalk in a time of day-dependent manner. Our findings highlight a crucial intersection between SC metabolic shifts and immune responses within the muscle microenvironment, dictated by the circadian rhythms, and underscore the potential for targeting circadian and metabolic pathways to enhance tissue regeneration.

2.
Nat Commun ; 15(1): 5046, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871717

RESUMEN

People with lower extremity peripheral artery disease (PAD) have increased oxidative stress, impaired mitochondrial activity, and poor walking performance. NAD+ reduces oxidative stress and is an essential cofactor for mitochondrial respiration. Oral nicotinamide riboside (NR) increases bioavailability of NAD+ in humans. Among 90 people with PAD, this randomized double-blind clinical trial assessed whether 6-months of NR, with and without resveratrol, improves 6-min walk distance, compared to placebo, at 6-month follow-up. At 6-month follow-up, compared to placebo, NR significantly improved 6-min walk (+7.0 vs. -10.6 meters, between group difference: +17.6 (90% CI: + 1.8,+∞). Among participants who took at least 75% of study pills, compared to placebo, NR improved 6-min walk by 31.0 meters and NR + resveratrol improved 6-min walk by 26.9 meters. In this work, NR meaningfully improved 6-min walk, and resveratrol did not add benefit to NR alone in PAD. A larger clinical trial to confirm these findings is needed.


Asunto(s)
Niacinamida , Enfermedad Arterial Periférica , Compuestos de Piridinio , Resveratrol , Humanos , Enfermedad Arterial Periférica/tratamiento farmacológico , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Masculino , Femenino , Anciano , Método Doble Ciego , Resveratrol/uso terapéutico , Resveratrol/farmacología , Persona de Mediana Edad , Caminata , Resultado del Tratamiento , Estrés Oxidativo/efectos de los fármacos
3.
J Clin Invest ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38722697

RESUMEN

Newborn mammalian cardiomyocytes quickly transition from a fetal to an adult phenotype that utilizes mitochondrial oxidative phosphorylation but loses mitotic capacity. We tested whether forced reversal of adult cardiomyocytes back to a fetal glycolytic phenotype would restore proliferative capacity. We deleted Uqcrfs1 (mitochondrial Rieske Iron-Sulfur protein, RISP) in hearts of adult mice. As RISP protein decreased, heart mitochondrial function declined, and glucose utilization increased. Simultaneously, they underwent hyperplastic remodeling during which cardiomyocyte number doubled without cellular hypertrophy. Cellular energy supply was preserved, AMPK activation was absent, and mTOR activation was evident. In ischemic hearts with RISP deletion, new cardiomyocytes migrated into the infarcted region, suggesting the potential for therapeutic cardiac regeneration. RNA-seq revealed upregulation of genes associated with cardiac development and proliferation. Metabolomic analysis revealed a decrease in alpha-ketoglutarate (required for TET-mediated demethylation) and an increase in S-adenosylmethionine (required for methyltransferase activity). Analysis revealed an increase in methylated CpGs near gene transcriptional start sites. Genes that were both differentially expressed and differentially methylated were linked to upregulated cardiac developmental pathways. We conclude that decreased mitochondrial function and increased glucose utilization can restore mitotic capacity in adult cardiomyocytes resulting in the generation of new heart cells, potentially through the modification of substrates that regulate epigenetic modification of genes required for proliferation.

4.
Curr Top Dev Biol ; 158: 307-339, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670711

RESUMEN

Recent research has highlighted an important role for the molecular circadian machinery in the regulation of tissue-specific function and stress responses. Indeed, disruption of circadian function, which is pervasive in modern society, is linked to accelerated aging, obesity, and type 2 diabetes. Furthermore, evidence supporting the importance of the circadian clock within both the mature muscle tissue and satellite cells to regulate the maintenance of muscle mass and repair capacity in response injury has recently emerged. Here, we review the discovery of circadian clocks within the satellite cell (a.k.a. adult muscle stem cell) and how they act to regulate metabolism, epigenetics, and myogenesis during both healthy and diseased states.


Asunto(s)
Ritmo Circadiano , Regeneración , Células Satélite del Músculo Esquelético , Células Satélite del Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Animales , Regeneración/fisiología , Humanos , Ritmo Circadiano/fisiología , Músculo Esquelético/fisiología , Desarrollo de Músculos , Relojes Circadianos/fisiología , Epigénesis Genética
5.
Life Sci Alliance ; 7(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38453366

RESUMEN

The recently discovered HAPSTR1 protein broadly oversees cellular stress responses. This function requires HUWE1, a ubiquitin ligase that paradoxically marks HAPSTR1 for degradation, but much about this pathway remains unclear. Here, leveraging multiplexed proteomics, we find that HAPSTR1 enables nuclear localization of HUWE1 with implications for nuclear protein quality control. We show that HAPSTR1 is tightly regulated and identify ubiquitin ligase TRIP12 and deubiquitinase USP7 as upstream regulators titrating HAPSTR1 stability. Finally, we generate conditional Hapstr1 knockout mice, finding that Hapstr1-null mice are perinatal lethal, adult mice depleted of Hapstr1 have reduced fitness, and primary cells explanted from Hapstr1-null animals falter in culture coincident with HUWE1 mislocalization and broadly remodeled signaling. Notably, although HAPSTR1 potently suppresses p53, we find that Hapstr1 is essential for life even in mice lacking p53. Altogether, we identify novel components and functional insights into the conserved HAPSTR1-HUWE1 pathway and demonstrate its requirement for mammalian life.


Asunto(s)
Proteína p53 Supresora de Tumor , Ubiquitina-Proteína Ligasas , Animales , Ratones , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/genética , Proteínas Nucleares/metabolismo , Transducción de Señal/genética , Mamíferos/metabolismo
6.
Mol Metab ; 62: 101528, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35717025

RESUMEN

OBJECTIVE: Mitochondrial capacity is critical to adapt the high energy demand of the heart to circadian oscillations and diseased states. Glucocorticoids regulate the circadian cycle of energy metabolism, but little is known about how circadian timing of exogenous glucocorticoid dosing directly regulates heart metabolism through cardiomyocyte-autonomous mechanisms. While chronic once-daily intake of glucocorticoids promotes metabolic stress and heart failure, we recently discovered that intermittent once-weekly dosing of exogenous glucocorticoids promoted muscle metabolism in normal and obese skeletal muscle. However, the effects of glucocorticoid intermittence on heart metabolism and heart failure remain unknown. Here we investigated the extent to which circadian time of dosing regulates the effects of the glucocorticoid prednisone in heart metabolism and function in conditions of single pulse or chronic intermittent dosing. METHODS AND RESULTS: In WT mice, we found that prednisone improved cardiac content of NAD+ and ATP with light-phase dosing (ZT0), while the effects were blocked by dark-phase dosing (ZT12). The drug effects on mitochondrial function were cardiomyocyte-autonomous, as shown by inducible cardiomyocyte-restricted glucocorticoid receptor (GR) ablation, and depended on an intact cardiomyocyte clock, as shown by inducible cardiomyocyte-restricted ablation of Brain and Muscle ARNT-like 1 (BMAL1). Conjugating time-of-dosing with chronic intermittence, we found that once-weekly prednisone improved metabolism and function in heart after myocardial injury dependent on circadian time of intake, i.e. with light-phase but not dark-phase dosing. CONCLUSIONS: Our study identifies cardiac-autonomous mechanisms through which circadian-specific intermittent dosing reconverts glucocorticoid drugs to metabolic boosters for the heart.


Asunto(s)
Relojes Circadianos , Insuficiencia Cardíaca , Animales , Relojes Circadianos/fisiología , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Insuficiencia Cardíaca/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Prednisona/metabolismo , Prednisona/farmacología
7.
Sci Adv ; 8(7): eabm1189, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35179955

RESUMEN

Exogenous glucocorticoids interact with the circadian clock, but little attention is paid to the timing of intake. We recently found that intermittent once-weekly prednisone improved nutrient oxidation in dystrophic muscle. Here, we investigated whether dosage time affected prednisone effects on muscle bioenergetics. In mice treated with once-weekly prednisone, drug dosing in the light-phase promoted nicotinamide adenine dinucleotide (NAD+) levels and mitochondrial function in wild-type muscle, while this response was lost with dark-phase dosing. These effects depended on a normal circadian clock since they were disrupted in muscle from [Brain and muscle Arnt-like protein-1 (Bmal1)]-knockout mice. The light-phase prednisone pulse promoted BMAL1-dependent glucocorticoid receptor recruitment on noncanonical targets, including Nampt and Ppargc1a [peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α)]. In mice with muscle-restricted inducible PGC1α ablation, bioenergetic stimulation by light-phase prednisone required PGC1α. These results demonstrate that glucocorticoid "chronopharmacology" for muscle bioenergetics requires an intact clock and muscle PGC1α activity.


Asunto(s)
Relojes Circadianos , Factores de Transcripción ARNTL/genética , Animales , Glucocorticoides/farmacología , Ratones , Mitocondrias/metabolismo , Músculos , NAD , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Prednisona
8.
Genes Dev ; 36(3-4): 149-166, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35115380

RESUMEN

The process of tissue regeneration occurs in a developmentally timed manner, yet the role of circadian timing is not understood. Here, we identify a role for the adult muscle stem cell (MuSC)-autonomous clock in the control of muscle regeneration following acute ischemic injury. We observed greater muscle repair capacity following injury during the active/wake period as compared with the inactive/rest period in mice, and loss of Bmal1 within MuSCs leads to impaired muscle regeneration. We demonstrate that Bmal1 loss in MuSCs leads to reduced activated MuSC number at day 3 postinjury, indicating a failure to properly expand the myogenic precursor pool. In cultured primary myoblasts, we observed that loss of Bmal1 impairs cell proliferation in hypoxia (a condition that occurs in the first 1-3 d following tissue injury in vivo), as well as subsequent myofiber differentiation. Loss of Bmal1 in both cultured myoblasts and in vivo activated MuSCs leads to reduced glycolysis and premature activation of prodifferentiation gene transcription and epigenetic remodeling. Finally, hypoxic cell proliferation and myofiber formation in Bmal1-deficient myoblasts are restored by increasing cytosolic NAD+ Together, we identify the MuSC clock as a pivotal regulator of oxygen-dependent myoblast cell fate and muscle repair through the control of the NAD+-driven response to injury.


Asunto(s)
Factores de Transcripción ARNTL , NAD , Células Satélite del Músculo Esquelético , Factores de Transcripción ARNTL/genética , Animales , Diferenciación Celular/genética , Hipoxia , Ratones , Desarrollo de Músculos/genética , Músculo Esquelético , Mioblastos
9.
Nat Metab ; 3(12): 1621-1632, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34903884

RESUMEN

In mammals, circadian rhythms are entrained to the light cycle and drive daily oscillations in levels of NAD+, a cosubstrate of the class III histone deacetylase sirtuin 1 (SIRT1) that associates with clock transcription factors. Although NAD+ also participates in redox reactions, the extent to which NAD(H) couples nutrient state with circadian transcriptional cycles remains unknown. Here we show that nocturnal animals subjected to time-restricted feeding of a calorie-restricted diet (TRF-CR) only during night-time display reduced body temperature and elevated hepatic NADH during daytime. Genetic uncoupling of nutrient state from NADH redox state through transduction of the water-forming NADH oxidase from Lactobacillus brevis (LbNOX) increases daytime body temperature and blood and liver acyl-carnitines. LbNOX expression in TRF-CR mice induces oxidative gene networks controlled by brain and muscle Arnt-like protein 1 (BMAL1) and peroxisome proliferator-activated receptor alpha (PPARα) and suppresses amino acid catabolic pathways. Enzymatic analyses reveal that NADH inhibits SIRT1 in vitro, corresponding with reduced deacetylation of SIRT1 substrates during TRF-CR in vivo. Remarkably, Sirt1 liver nullizygous animals subjected to TRF-CR display persistent hypothermia even when NADH is oxidized by LbNOX. Our findings reveal that the hepatic NADH cycle links nutrient state to whole-body energetics through the rhythmic regulation of SIRT1.


Asunto(s)
Metabolismo Energético , Ayuno , NAD/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Transcripción Genética , Aminoácidos/metabolismo , Animales , Temperatura Corporal , Ritmo Circadiano , Dieta , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Hígado/metabolismo , Ratones , Factores de Transcripción
10.
Exp Gerontol ; 140: 111048, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32755612

RESUMEN

OBJECTIVE: This study investigated associations of markers of oxidative stress and mitochondrial function in calf muscle biopsies with walking performance in people with and without lower extremity peripheral artery disease (PAD). METHODS: Participants with PAD (ankle-brachial index (ABI) <0.90) and without PAD (ABI: 0.90-1.50) underwent calf muscle biopsy and measurement of 6-min walk and four-meter walking velocity. PARP1 (Poly (ADP-Ribose) Polymerase 1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), silent information regulator 1 (SIRT1) and 4-hydroxynonenal (4HNE) expression were measured in calf muscle using western blot. RESULTS: Among 15 participants with PAD mean age: 66.8 years (standard deviation (SD): 6.4) and six without PAD (age: 64.4 years, SD: 5.9), mean PARP1-abundance in calf muscle was 1.16 ± 0.92 AU and 0.96 ± 0.38 AU, respectively (P = 0.61). Among participants with PAD after adjustment with ABI, a greater abundance of PARP1 was associated with poorer 6-min walking distance (r = -0.65, P = 0.01), usual-paced 4-m walking velocity (r = -0.73, P = 0.003) and slower fast-paced four-meter walking velocity (r = -0.51, P = 0.07). Among participants with PAD, ABI was not associated with PARP1 abundance in calf muscle (r = 0.02, P = 0.93). Among participants without PAD, skeletal muscle PARP1 abundance was not significantly associated with 6-min walk distance (r = -0.58; P = 0.22), usual-paced walking velocity (r = -0.26; P = 0.62), or fast-paced walking velocity (r = -0.21; P = 0.69), perhaps due to lack of statistical power. There were no associations of remaining calf muscle measures with walking performance. CONCLUSIONS: These findings are consistent with the hypothesis that calf skeletal muscle characteristics are related to walking performance, independently of severity of lower extremity arterial obstruction in people with PAD.


Asunto(s)
Enfermedad Arterial Periférica , Ribosa , Adenosina Difosfato , Anciano , Humanos , Músculo Esquelético , Poli Adenosina Difosfato Ribosa , Caminata
11.
Mol Cell ; 78(5): 835-849.e7, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32369735

RESUMEN

Disrupted sleep-wake and molecular circadian rhythms are a feature of aging associated with metabolic disease and reduced levels of NAD+, yet whether changes in nucleotide metabolism control circadian behavioral and genomic rhythms remains unknown. Here, we reveal that supplementation with the NAD+ precursor nicotinamide riboside (NR) markedly reprograms metabolic and stress-response pathways that decline with aging through inhibition of the clock repressor PER2. NR enhances BMAL1 chromatin binding genome-wide through PER2K680 deacetylation, which in turn primes PER2 phosphorylation within a domain that controls nuclear transport and stability and that is mutated in human advanced sleep phase syndrome. In old mice, dampened BMAL1 chromatin binding, transcriptional oscillations, mitochondrial respiration rhythms, and late evening activity are restored by NAD+ repletion to youthful levels with NR. These results reveal effects of NAD+ on metabolism and the circadian system with aging through the spatiotemporal control of the molecular clock.


Asunto(s)
Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Proteínas Circadianas Period/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Edad , Envejecimiento/genética , Animales , Proteínas CLOCK/genética , Ritmo Circadiano/fisiología , Citocinas/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , NAD/metabolismo , Proteínas Circadianas Period/genética , Sirtuina 1/metabolismo , Sirtuinas/metabolismo
12.
Trends Endocrinol Metab ; 31(6): 459-468, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32396846

RESUMEN

Research over the past few decades has shed light on the mechanisms underlying the link between circadian disruption and the development of metabolic diseases such as obesity, type 2 diabetes, and cancer. However, how the clock network interacts with tissue-specificnutrient-sensing pathways during conditions of nutrient stress or pathological states remains incompletely understood. Recent work has demonstrated that the circadian clock can 'reprogram' the transcriptome to control distinct sets of genes during altered nutrient conditions, such as high fat diet, aging, and exercise. In this review, I discuss connections between circadian clock transcription factors and the oxygen- and nutrient-responsivehypoxia-inducible factor (HIF) pathway. I highlight recently uncovered mechanistic insights underlying these pathway interactions and address potential implications for the role of circadian disruption in metabolic diseases.


Asunto(s)
Relojes Circadianos/fisiología , Regulación de la Expresión Génica/fisiología , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Oxígeno/metabolismo , Animales , Relojes Circadianos/genética , Humanos
13.
JCI Insight ; 4(24)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31852847

RESUMEN

In humans, chronic glucocorticoid use is associated with side effects like muscle wasting, obesity, and metabolic syndrome. Intermittent steroid dosing has been proposed in Duchenne Muscular Dystrophy patients to mitigate the side effects seen with daily steroid intake. We evaluated biomarkers from Duchenne Muscular Dystrophy patients, finding that, compared with chronic daily steroid use, weekend steroid use was associated with reduced serum insulin, free fatty acids, and branched chain amino acids, as well as reduction in fat mass despite having similar BMIs. We reasoned that intermittent prednisone administration in dystrophic mice would alter muscle epigenomic signatures, and we identified the coordinated action of the glucocorticoid receptor, KLF15 and MEF2C as mediators of a gene expression program driving metabolic reprogramming and enhanced nutrient utilization. Muscle lacking Klf15 failed to respond to intermittent steroids. Furthermore, coadministration of the histone acetyltransferase inhibitor anacardic acid with steroids in mdx mice eliminated steroid-specific epigenetic marks and abrogated the steroid response. Together, these findings indicate that intermittent, repeated exposure to glucocorticoids promotes performance in dystrophic muscle through an epigenetic program that enhances nutrient utilization.


Asunto(s)
Glucocorticoides/administración & dosificación , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Prednisona/administración & dosificación , Ácidos Anacárdicos/administración & dosificación , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Niño , Estudios Transversales , Modelos Animales de Enfermedad , Quimioterapia Combinada , Epigénesis Genética/efectos de los fármacos , Epigenómica , Regulación de la Expresión Génica/efectos de los fármacos , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción MEF2/metabolismo , Masculino , Metabolómica , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/genética , Nutrientes/sangre , Nutrientes/metabolismo , Quimioterapia por Pulso
14.
Curr Biol ; 29(12): 1954-1962.e4, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31155351

RESUMEN

In plants, cryptochromes are photoreceptors that negatively regulate the ubiquitin ligase CRL4Cop1. In mammals, cryptochromes are core components of the circadian clock and repressors of the glucocorticoid receptor (GR). Moreover, mammalian cryptochromes lost their ability to interact with Cop1, suggesting that they are unable to inhibit CRL4Cop1. Contrary to this assumption, we found that mammalian cryptochromes are also negative regulators of CRL4Cop1, and through this mechanism, they repress the GR transcriptional network both in cultured cells and in the mouse liver. Mechanistically, cryptochromes inactivate Cop1 by interacting with Det1, a subunit of the mammalian CRL4Cop1 complex that is not present in other CRL4s. Through this interaction, the ability of Cop1 to join the CRL4 complex is inhibited; therefore, its substrates accumulate. Thus, the interaction between cryptochromes and Det1 in mammals mirrors the interaction between cryptochromes and Cop1 in planta, pointing to a common ancestor in which the cryptochromes-Cop1 axis originated.


Asunto(s)
Criptocromos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Animales , Evolución Biológica , Línea Celular , Femenino , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
Cell Metab ; 29(5): 1078-1091.e5, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827863

RESUMEN

The alignment of fasting and feeding with the sleep/wake cycle is coordinated by hypothalamic neurons, though the underlying molecular programs remain incompletely understood. Here, we demonstrate that the clock transcription pathway maximizes eating during wakefulness and glucose production during sleep through autonomous circadian regulation of NPY/AgRP neurons. Tandem profiling of whole-cell and ribosome-bound mRNAs in morning and evening under dynamic fasting and fed conditions identified temporal control of activity-dependent gene repertoires in AgRP neurons central to synaptogenesis, bioenergetics, and neurotransmitter and peptidergic signaling. Synaptic and circadian pathways were specific to whole-cell RNA analyses, while bioenergetic pathways were selectively enriched in the ribosome-bound transcriptome. Finally, we demonstrate that the AgRP clock mediates the transcriptional response to leptin. Our results reveal that time-of-day restriction in transcriptional control of energy-sensing neurons underlies the alignment of hunger and food acquisition with the sleep/wake state.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Relojes Circadianos/genética , Ritmo Circadiano/genética , Hambre/fisiología , Neuronas/metabolismo , Transcripción Genética/genética , Proteína Relacionada con Agouti/genética , Animales , Ingestión de Alimentos/fisiología , Ayuno/fisiología , Redes Reguladoras de Genes , Glucosa/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/genética , Sueño/fisiología , Transcriptoma , Vigilia/fisiología
16.
Nat Commun ; 9(1): 3659, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30201971

RESUMEN

The HMG-CoA reductase degradation protein 1 (HRD1) has been identified as a key enzyme for endoplasmic reticulum-associated degradation of misfolded proteins, but its organ-specific physiological functions remain largely undefined. Here we show that mice with HRD1 deletion specifically in the liver display increased energy expenditure and are resistant to HFD-induced obesity and liver steatosis and insulin resistance. Proteomic analysis identifies a HRD1 interactome, a large portion of which includes metabolic regulators. Loss of HRD1 results in elevated ENTPD5, CPT2, RMND1, and HSD17B4 protein levels and a consequent hyperactivation of both AMPK and AKT pathways. Genome-wide mRNA sequencing revealed that HRD1-deficiency reprograms liver metabolic gene expression profiles, including suppressing genes involved in glycogenesis and lipogenesis and upregulating genes involved in glycolysis and fatty acid oxidation. We propose HRD1 as a liver metabolic regulator and a potential drug target for obesity, fatty liver disease, and insulin resistance associated with the metabolic syndrome.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Hígado/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adenilato Quinasa/metabolismo , Animales , Peso Corporal , Dieta Alta en Grasa , Activación Enzimática , Ácidos Grasos/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Glucólisis , Células HEK293 , Células Hep G2 , Humanos , Lipogénesis , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Proteoma , Proteómica , Triglicéridos/metabolismo , Ubiquitinación
17.
Cell Metab ; 25(1): 86-92, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-27773696

RESUMEN

Circadian clocks are encoded by a transcription-translation feedback loop that aligns energetic processes with the solar cycle. We show that genetic disruption of the clock activator BMAL1 in skeletal myotubes and fibroblasts increased levels of the hypoxia-inducible factor 1α (HIF1α) under hypoxic conditions. Bmal1-/- myotubes displayed reduced anaerobic glycolysis, mitochondrial respiration with glycolytic fuel, and transcription of HIF1α targets Phd3, Vegfa, Mct4, Pk-m, and Ldha, whereas abrogation of the clock repressors CRY1/2 stabilized HIF1α in response to hypoxia. HIF1α bound directly to core clock gene promoters, and, when co-expressed with BMAL1, led to transactivation of PER2-LUC and HRE-LUC reporters. Further, genetic stabilization of HIF1α in Vhl-/- cells altered circadian transcription. Finally, induction of clock and HIF1α target genes in response to strenuous exercise varied according to the time of day in wild-type mice. Collectively, our results reveal bidirectional interactions between circadian and HIF pathways that influence metabolic adaptation to hypoxia.


Asunto(s)
Relojes Circadianos , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Músculo Esquelético/metabolismo , Oxígeno/metabolismo , Anaerobiosis , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Hipoxia/genética , Hipoxia/metabolismo , Ratones , Especificidad de Órganos , Consumo de Oxígeno , Condicionamiento Físico Animal , Transcripción Genética
18.
Ann N Y Acad Sci ; 1385(1): 21-40, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27589593

RESUMEN

This paper focuses on the relationship between the circadian system and glucose metabolism. Research across the translational spectrum confirms the importance of the circadian system for glucose metabolism and offers promising clues as to when and why these systems go awry. In particular, basic research has started to clarify the molecular and genetic mechanisms through which the circadian system regulates metabolism. The study of human behavior, especially in the context of psychiatric disorders, such as bipolar disorder and major depression, forces us to see how inextricably linked mental health and metabolic health are. We also emphasize the remarkable opportunities for advancing circadian science through big data and advanced analytics. Advances in circadian research have translated into environmental and pharmacological interventions with tremendous therapeutic potential.


Asunto(s)
Encéfalo/metabolismo , Ritmo Circadiano/fisiología , Tracto Gastrointestinal/metabolismo , Enfermedades Metabólicas/metabolismo , Trastornos del Humor/metabolismo , Animales , Ensayos Clínicos como Asunto/métodos , Humanos , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/psicología , Trastornos del Humor/diagnóstico , Trastornos del Humor/psicología
19.
Science ; 350(6261): aac4250, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26542580

RESUMEN

The mammalian transcription factors CLOCK and BMAL1 are essential components of the molecular clock that coordinate behavior and metabolism with the solar cycle. Genetic or environmental perturbation of circadian cycles contributes to metabolic disorders including type 2 diabetes. To study the impact of the cell-autonomous clock on pancreatic ß cell function, we examined pancreatic islets from mice with either intact or disrupted BMAL1 expression both throughout life and limited to adulthood. We found pronounced oscillation of insulin secretion that was synchronized with the expression of genes encoding secretory machinery and signaling factors that regulate insulin release. CLOCK/BMAL1 colocalized with the pancreatic transcription factor PDX1 within active enhancers distinct from those controlling rhythmic metabolic gene networks in liver. We also found that ß cell clock ablation in adult mice caused severe glucose intolerance. Thus, cell type-specific enhancers underlie the circadian control of peripheral metabolism throughout life and may help to explain its dysregulation in diabetes.


Asunto(s)
Ritmo Circadiano/genética , Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Exocitosis/genética , Intolerancia a la Glucosa , Proteínas de Homeodominio/metabolismo , Humanos , Secreción de Insulina , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transactivadores/metabolismo , Transcripción Genética
20.
Science ; 342(6158): 1243417, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24051248

RESUMEN

Circadian clocks are self-sustained cellular oscillators that synchronize oxidative and reductive cycles in anticipation of the solar cycle. We found that the clock transcription feedback loop produces cycles of nicotinamide adenine dinucleotide (NAD(+)) biosynthesis, adenosine triphosphate production, and mitochondrial respiration through modulation of mitochondrial protein acetylation to synchronize oxidative metabolic pathways with the 24-hour fasting and feeding cycle. Circadian control of the activity of the NAD(+)-dependent deacetylase sirtuin 3 (SIRT3) generated rhythms in the acetylation and activity of oxidative enzymes and respiration in isolated mitochondria, and NAD(+) supplementation restored protein deacetylation and enhanced oxygen consumption in circadian mutant mice. Thus, circadian control of NAD(+) bioavailability modulates mitochondrial oxidative function and organismal metabolism across the daily cycles of fasting and feeding.


Asunto(s)
Relojes Circadianos/fisiología , Metabolismo Energético , Mitocondrias Hepáticas/metabolismo , NAD/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Acetilación , Animales , Relojes Circadianos/genética , Ayuno , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Noqueados , Oxidación-Reducción , Consumo de Oxígeno , Sirtuina 3/genética , Sirtuina 3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...