Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 159(2): 306-17, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303527

RESUMEN

Induction of beige cells causes the browning of white fat and improves energy metabolism. However, the central mechanism that controls adipose tissue browning and its physiological relevance are largely unknown. Here, we demonstrate that fasting and chemical-genetic activation of orexigenic AgRP neurons in the hypothalamus suppress the browning of white fat. O-linked ß-N-acetylglucosamine (O-GlcNAc) modification of cytoplasmic and nuclear proteins regulates fundamental cellular processes. The levels of O-GlcNAc transferase (OGT) and O-GlcNAc modification are enriched in AgRP neurons and are elevated by fasting. Genetic ablation of OGT in AgRP neurons inhibits neuronal excitability through the voltage-dependent potassium channel, promotes white adipose tissue browning, and protects mice against diet-induced obesity and insulin resistance. These data reveal adipose tissue browning as a highly dynamic physiological process under central control, in which O-GlcNAc signaling in AgRP neurons is essential for suppressing thermogenesis to conserve energy in response to fasting.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Dieta , N-Acetilglucosaminiltransferasas/metabolismo , Neuronas/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Ayuno , Femenino , Ghrelina/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , N-Acetilglucosaminiltransferasas/genética , Obesidad/metabolismo , Obesidad/prevención & control
2.
Yi Chuan ; 45(2): 144-155, 2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36927661

RESUMEN

Neural regulation of adipose tissue is crucial in the homeostasis of energy metabolism. Adipose tissue neuropeptide Y (NPY) and its receptors contribute to the development of diet-induced obesity. NPY1R and NPY2R are major receptors for NPY in peripheral tissues including the adipose tissue. NPY receptor 4 (Npy4r) gene is expressed in adipose tissue. However, it is unknown whether Npy4r is involved in the development of diet-induced obesity. Here, we established an immunofluorescence microscopy technique and generated an adipocyte-reconstituted Npy4r gene knockout mouse. Among six adipose depots, we found that NPY is highly expressed around the vasculature in a dot-like fashion in interscapular brown fat and subcutaneous fat, and NPY receptors are expressed in a depot-specific manner. NPY1R is highly expressed in epidydimal fat, interscapular and peri-aortic brown fat, NPY2R in both interscapular and peri-aortic brown fat, and NPY4R in both brown fat and epidydimal fat. Next, we showed that adipocyte-reconstituted expression of Npy4r promoted diet-induced obesity in mice (P < 0.0001). Overall, this study defines the abundance and distribution of NPY and its receptors 1, 2, and 4 in mouse adipose depots, and demonstrates in an adipocyte-reconstituted gene knockout model that adipocyte Npy4r is sufficient to promote diet-induced obesity.


Asunto(s)
Adipocitos , Obesidad , Ratones , Animales , Obesidad/genética , Adipocitos/metabolismo , Dieta , Tejido Adiposo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo
3.
Sheng Li Xue Bao ; 74(5): 726-736, 2022 Oct 25.
Artículo en Zh | MEDLINE | ID: mdl-36319096

RESUMEN

The central circadian clock and feeding rhythm coordinately reset peripheral circadian clocks. Emerging evidence suggests that feeding rhythm resets peripheral circadian clocks in a tissue-specific manner. This study aimed to determine whether and how feeding rhythm regulates circadian rhythms of the circadian clock and metabolic genes in brown adipose tissue (BAT). We applied different regimens of time-restricted feeding (TRF) in wildtype and Per1/2 deficient C57BL/6 mice, and quantified the effects of sex, treatment duration, constant light, and circadian clock on circadian rhythms of the BAT circadian clock and metabolic genes by RT-qPCR; Representative circadian clock genes are Bmal1, Nr1d1, Dbp, and Per2, and representative metabolic genes are uncoupling protein 1 (Ucp1), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3) that controls the flux through glycolysis, pyruvate dehydrogenase kinase isozyme 4 (Pdk4) gating the tricarboxylic acid cycle, and carnitine palmitoyltransferase 1A (Cpt1a) that controls mitochondrial fatty acid oxidation. The results showed that, daytime-restricted feeding (DRF) moderately shifted the phase of the BAT circadian clock in female mice within 7 or 36 d, and resulted in the loss of circadian rhythm in Dbp and Per2 transcripts in males. DRF induced de novo oscillation of the Ucp1 transcript, and shifted the phase of representative metabolic genes, such as Pfkfb3, Pdk4, and Cpt1a, more than 7 h. Constant light is known to disrupt the synchrony of the central circadian clock. The results showed that constant light promoted phase entrainment of the circadian clock by DRF in BAT, but abolished the oscillation of the metabolic genes (except for Pdk4). Despite combined treatment with Per1/2 deficiency and constant darkness, DRF was sufficient to drive circadian rhythms of Bmal1 and Dbp, but not those of Nr1d1, Ucp1, Pfkfb3, and Cpt1a. Overall, the circadian clock of BAT has weak adaptation to altered feeding rhythms and sex differences. The central circadian clock antagonizes DRF in the entrainment of the BAT circadian clock, whereas DRF resets circadian rhythms of metabolic genes, such as Ucp1, Pfkfb3, and Cpt1a, in a circadian clock-dependent manner.


Asunto(s)
Relojes Circadianos , Femenino , Masculino , Animales , Ratones , Ratones Endogámicos C57BL , Tejido Adiposo Pardo , Factores de Transcripción ARNTL , Ritmo Circadiano
4.
Yi Chuan ; 44(10): 950-957, 2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36384730

RESUMEN

Adipose tissue plays an important role in metabolic physiology through energy storage and endocrine functions. Spatial transcriptomics is revealing the complexity of cell types and their interaction in the adipose tissue with regards to development, homeostasis and disease. Emerging evidence suggests the existence of different subtypes of mature adipocytes that may have distinct functions, the markers of which include leptin (LEP), adiponectin (ADIPOQ), perilipin-1/4 (PLIN), and serum amyloid A (SAA), marking different adipocyte subtypes. Currently, Adipoq-Cre is widely used to study adipocyte biology, however, there is no Cre line that specifically targets LEP+ adipocytes. Here, we report the construction and validation of a Lep-Cre mouse line, which has the endogenous Lep gene edited by the CRISPR-Cas9 technology to generate the Lep-peptide 2A (P2A)-Cre fusion gene. P2A induces an auto-hydrolysis of the fusion protein, leading to expression of the Cre recombinase by the Lep gene activity. The activity of Lep-Cre in different depots of adipose tissues and non-adipose tissues was visualized by the immunofluorescence microscopy in the Lep-Cre Rosa26-loxP-Stop-loxP-tdTomato mice. We showed that Lep-Cre marked white/beige adipose depots extensively, followed by brown adipose depots. Leaky activity was observed in varying degrees among peripheral organs but not in the paraventricular nucleus of the hypothalamus. In summary, we have constructed a new adipocyte-targeting Cre mouse line that would be useful to study the development and physiology of LEP+ adipocytes.


Asunto(s)
Adipocitos , Leptina , Ratones , Animales , Leptina/genética , Leptina/metabolismo , Adipocitos/metabolismo , Integrasas/genética , Integrasas/metabolismo , Transgenes
5.
J Biol Chem ; 294(25): 9720-9721, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227622

RESUMEN

Adropin is a liver-secreted peptide that is crucial for metabolic health. However, the molecular functions and clinical significance of adropin have not been adequately explored. Butler et al. now investigate adropin expression profiles and links to cardiometabolic disease risk in two nonhuman primate models, increasing our translational and mechanistic understanding of this fascinating hormone.


Asunto(s)
Péptidos , Aumento de Peso , Animales , Dieta , Masculino , Primates , Azúcares
6.
J Biol Chem ; 293(36): 13989-14000, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30037904

RESUMEN

Many intracellular proteins are reversibly modified by O-linked GlcNAc (O-GlcNAc), a post-translational modification that dynamically regulates fundamental cellular processes in response to diverse environmental cues. Accumulating evidence indicates that both excess and deficiency of protein O-GlcNAcylation can have deleterious effects on the cell, suggesting that maintenance of O-GlcNAc homeostasis is essential for proper cellular function. However, the mechanisms through which O-GlcNAc homeostasis is maintained in the physiologic state and altered in the disease state have not yet been investigated. Here, we demonstrate the existence of a homeostatic mechanism involving mutual regulation of the O-GlcNAc-cycling enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) at the transcriptional level. Specifically, we found that OGA promotes Ogt transcription through cooperation with the histone acetyltransferase p300 and transcription factor CCAAT/enhancer-binding protein ß (C/EBPß). To examine the role of mutual regulation of OGT and OGA in the disease state, we analyzed gene expression data from human cancer data sets, which revealed that OGT and OGA expression levels are highly correlated in numerous human cancers, particularly in pancreatic adenocarcinoma. Using a KrasG12D -driven primary mouse pancreatic ductal adenocarcinoma (PDAC) cell line, we found that inhibition of extracellular signal-regulated kinase (ERK) signaling decreases OGA glycosidase activity and reduces OGT mRNA and protein levels, suggesting that ERK signaling may alter O-GlcNAc homeostasis in PDAC by modulating OGA-mediated Ogt transcription. Our study elucidates a transcriptional mechanism that regulates cellular O-GlcNAc homeostasis, which may lay a foundation for exploring O-GlcNAc signaling as a therapeutic target for human disease.


Asunto(s)
Acetilglucosamina/metabolismo , Regulación Neoplásica de la Expresión Génica , Homeostasis , Neoplasias Pancreáticas/metabolismo , Animales , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Glicósido Hidrolasas , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , N-Acetilglucosaminiltransferasas , Neoplasias Pancreáticas/genética , Procesamiento Proteico-Postraduccional , Transducción de Señal
7.
J Biol Chem ; 294(30): 11653, 2019 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-31350285
8.
J Biol Chem ; 287(16): 12904-12, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22371499

RESUMEN

Recruitment of O-GlcNAc transferase (OGT) to promoters plays an important role in gene repression. Glucocorticoid signaling represses the transcriptional activities of NF-κB and AP-1 through direct binding, yet the molecular mechanisms remain to be elucidated. Here we report that OGT is an important component of GR-mediated transrepression. OGT associates with ligand-bound GR in a multi-protein repression complex. Overexpression of OGT potentiates the GR transrepression pathway, whereas depletion of endogenous OGT by RNA interference abolishes the repression. The recruitment of OGT by GR leads to increased O-GlcNAcylation and decreased phosphorylation of RNA polymerase II on target genes. Functionally, overexpression of OGT enhances glucocorticoid-induced apoptosis in resistant cell lines while knockdown of OGT prevents sensitive cell lines from apoptosis. These studies identify a molecular mechanism of GR transrepression, and highlight the function of O-GlcNAc in hormone signaling.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Acetilglucosamina/metabolismo , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Apoptosis/fisiología , Línea Celular Tumoral , Diseño de Fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Hidrocortisona/metabolismo , Hidrocortisona/farmacología , Neoplasias Pulmonares , FN-kappa B/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , ARN Polimerasa II/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología
9.
Nat Commun ; 14(1): 6086, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773240

RESUMEN

Post-translational modifications (PTMs) couple feed-fast cycles to diurnal rhythms. However, it remains largely uncharacterized whether and how meal timing organizes diurnal rhythms beyond the transcriptome. Here, we systematically profile the daily rhythms of the proteome, four PTMs (phosphorylation, ubiquitylation, succinylation and N-glycosylation) and the lipidome in the liver from young female mice subjected to either day/sleep time-restricted feeding (DRF) or night/wake time-restricted feeding (NRF). We detect robust daily rhythms among different layers of omics with phosphorylation the most nutrient-responsive and succinylation the least. Integrative analyses reveal that clock regulation of fatty acid metabolism represents a key diurnal feature that is reset by meal timing, as indicated by the rhythmic phosphorylation of the circadian repressor PERIOD2 at Ser971 (PER2-pSer971). We confirm that PER2-pSer971 is activated by nutrient availability in vivo. Together, this dataset represents a comprehensive resource detailing the proteomic and lipidomic responses by the liver to alterations in meal timing.


Asunto(s)
Relojes Circadianos , Multiómica , Femenino , Ratones , Animales , Proteómica , Ritmo Circadiano/fisiología , Sueño , Hígado/metabolismo , Relojes Circadianos/fisiología
10.
Nat Metab ; 5(7): 1236-1251, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37365376

RESUMEN

Physical endurance and energy conservation are essential for survival in the wild. However, it remains unknown whether and how meal timing regulates physical endurance and muscle diurnal rhythms. Here, we show that day/sleep time-restricted feeding (DRF) enhances running endurance by 100% throughout the circadian cycle in both male and female mice, compared to either ad libitum feeding or night/wake time-restricted feeding. Ablation of the circadian clock in the whole body or the muscle abolished the exercise regulatory effect of DRF. Multi-omics analysis revealed that DRF robustly entrains diurnal rhythms of a mitochondrial oxidative metabolism-centric network, compared to night/wake time-restricted feeding. Remarkably, muscle-specific knockdown of the myocyte lipid droplet protein perilipin-5 completely mimics DRF in enhancing endurance, enhancing oxidative bioenergetics and outputting rhythmicity to circulating energy substrates, including acylcarnitine. Together, our work identifies a potent dietary regimen to enhance running endurance without prior exercise, as well as providing a multi-omics atlas of muscle circadian biology regulated by meal timing.


Asunto(s)
Relojes Circadianos , Carrera , Femenino , Ratones , Masculino , Animales , Ritmo Circadiano/fisiología , Relojes Circadianos/fisiología
11.
Yale J Biol Med ; 85(3): 387-401, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23012586

RESUMEN

The circadian clock is a highly conserved timing system, resonating physiological processes to 24-hour environmental cycles. Circadian misalignment is emerging as a risk factor of metabolic disease. The molecular clock resides in all metabolic tissues, the dysfunction of which is associated with perturbed energy metabolism. In this article, we will review current knowledge about molecular mechanisms of the circadian clock and the role of clocks in the physiology and pathophysiology of metabolic tissues.


Asunto(s)
Relojes Circadianos , Metabolismo Energético , Enfermedades Metabólicas/fisiopatología , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Temperatura Corporal , Glucosa/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/fisiopatología , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Páncreas/metabolismo , Páncreas/fisiopatología , Factores de Riesgo
12.
Trends Mol Med ; 28(1): 25-35, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34801412

RESUMEN

Time-restricted eating (TRE), which limits the daily meal timing to a window of 6-12 h, has been shown to reduce the risks of cardiometabolic diseases through consolidating circadian rhythms of metabolism and physiology. Recent advances indicate that canonical circadian clocks are dispensable for the actions of TRE in the liver, and that meal timing entrains circadian rhythms in peripheral tissues in a tissue-specific manner (e.g., the liver and fat are readily entrainable, whereas the heart and kidneys are resistant). Here, we propose that TRE engages clock-modulated checkpoints (CCPs) to reset circadian rhythms of tissue functions. Elucidation of CCPs would reveal the mechanistic basis of tissue responsiveness to TRE, and facilitate the use of TRE in precision medicine for cardiometabolic diseases.


Asunto(s)
Relojes Circadianos , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Ingestión de Alimentos , Humanos , Hígado
13.
J Mol Cell Biol ; 14(3)2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35285892

RESUMEN

O-linked N-acetyl-glucosamine glycosylation (O-GlcNAcylation) of intracellular proteins is a dynamic process broadly implicated in age-related disease, yet it remains uncharacterized whether and how O-GlcNAcylation contributes to the natural aging process. O-GlcNAc transferase (OGT) and the opposing enzyme O-GlcNAcase (OGA) control this nutrient-sensing protein modification in cells. Here, we show that global O-GlcNAc levels are increased in multiple tissues of aged mice. In aged liver, carbamoyl phosphate synthetase 1 (CPS1) is among the most heavily O-GlcNAcylated proteins. CPS1 O-GlcNAcylation is reversed by calorie restriction and is sensitive to genetic and pharmacological manipulations of the O-GlcNAc pathway. High glucose stimulates CPS1 O-GlcNAcylation and inhibits CPS1 activity. Liver-specific deletion of OGT potentiates CPS1 activity and renders CPS1 irresponsive to further stimulation by a prolonged fasting. Our results identify CPS1 O-GlcNAcylation as a key nutrient-sensing regulatory step in the urea cycle during aging and dietary restriction, implying a role for mitochondrial O-GlcNAcylation in nutritional regulation of longevity.


Asunto(s)
Hígado , Procesamiento Proteico-Postraduccional , Acetilglucosamina/metabolismo , Envejecimiento , Animales , Glicosilación , Hígado/metabolismo , Ratones , Urea/metabolismo
14.
F1000Res ; 11: 1087, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36531263

RESUMEN

Background: Meal timing resets circadian clocks in peripheral tissues, such as the liver, in seven days without affecting the phase of the central clock located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Anterior hypothalamus plays an essential role in energy metabolism, circadian rhythm, and stress response. However, it remains to be elucidated whether and how anterior hypothalamus adapts its circadian rhythms to meal timing. Methods: Here, we applied transcriptomics to profile rhythmic transcripts in the anterior hypothalamus of nocturnal female mice subjected to day- (DRF) or night (NRF)-time restricted feeding for seven days. Results: This global profiling identified 128 and 3,518 rhythmic transcripts in DRF and NRF, respectively. NRF entrained diurnal rhythms among 990 biological processes, including 'Electron transport chain' and 'Hippo signaling' that reached peak time in the late sleep and late active phase, respectively. By contrast, DRF entrained only 20 rhythmic pathways, including 'Cellular amino acid catabolic process', all of which were restricted to the late active phase. The rhythmic transcripts found in both DRF and NRF tissues were largely resistant to phase entrainment by meal timing, which were matched to the action of the circadian clock. Remarkably, DRF for 36 days partially reversed the circadian clock compared to NRF. Conclusions: Collectively, our work generates a useful dataset to explore anterior hypothalamic circadian biology and sheds light on potential rhythmic processes influenced by meal timing in the brain (www.circametdb.org.cn).


Asunto(s)
Relojes Circadianos , Núcleo Supraquiasmático , Femenino , Animales , Ratones , Núcleo Supraquiasmático/metabolismo , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Hipotálamo , Hígado
15.
Front Physiol ; 13: 953237, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36117681

RESUMEN

High-fat diet (HFD) feeding rewires circadian rhythms of peripheral organs including the liver and adipose tissue. While the liver has been extensively studied, it remains largely unknown whether and how HFD organizes circadian biology in adipose tissue. Here, we took a systems approach to profile the diurnal transcriptome of adipose tissue in diet-induced obese mice either fed a low-fat diet (LFD) that reduces weight or still fed HFD. We detected about 200 and 2,500 diurnal genes in HFD and LFD, respectively. Pathway analysis revealed that rhythmic pathways in HFD are represented by circadian rhythm, ribosome biogenesis, and nucleosome organization, whereas those in LFD are represented by myeloid cell function. Remarkably, the majority of the circadian clock genes, except Clock, exhibited robust diurnal rhythm in the adipose tissue of HFD-fed mice. Analysis of mRNAs and proteins in another cohort of HFD-fed mice confirmed that Clock lost rhythmicity at the transcript, but not protein level. Diet reversal to LFD specifically restored diurnal difference of the Clock transcripts in adipose tissue. We matched transcriptomics data with global profiling of neutral lipids and found that lipid metabolism catalyzed by triglycerol hydrolase Ces1d is a key circadian feature that is activated by diet reversal. Together, our work defines the circadian signatures in the adipose tissue of diet-induced obese mice, and their flexibility upon dietary intervention, thereby shedding light on potential clock-modulated tissue-specific pathways during obesity.

16.
J Mol Cell Biol ; 14(8)2022 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-36107452

RESUMEN

Comparative gene identification-58 (CGI-58), also known as α/ß hydrolase domain containing 5, is the co-activator of adipose triglyceride lipase that hydrolyzes triglycerides stored in the cytosolic lipid droplets. Mutations in CGI-58 gene cause Chanarin-Dorfman syndrome (CDS), an autosomal recessive neutral lipid storage disease with ichthyosis. The liver pathology of CDS manifests as steatosis and steatohepatitis, which currently has no effective treatments. Perilipin-3 (Plin3) is a member of the Perilipin-ADRP-TIP47 protein family that is essential for lipid droplet biogenesis. The objective of this study was to test a hypothesis that deletion of a major lipid droplet protein alleviates fatty liver pathogenesis caused by CGI-58 deficiency in hepatocytes. Adult CGI-58-floxed mice were injected with adeno-associated vectors simultaneously expressing the Cre recombinase and microRNA against Plin3 under the control of a hepatocyte-specific promoter, followed by high-fat diet feeding for 6 weeks. Liver and blood samples were then collected from these animals for histological and biochemical analysis. Plin3 knockdown in hepatocytes prevented steatosis, steatohepatitis, and necroptosis caused by hepatocyte CGI-58 deficiency. Our work is the first to show that inhibiting Plin3 in hepatocytes is sufficient to mitigate hepatocyte CGI-58 deficiency-induced hepatic steatosis and steatohepatitis in mice.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa , Hígado Graso , Ratones , Animales , Perilipina-3 , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Triglicéridos/metabolismo
17.
Yale J Biol Med ; 84(1): 1-7, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21451778

RESUMEN

The 2010 Lasker Award for basic medical research was shared by Douglas Coleman and Jeffery Friedman for their discovery of leptin, a breakthrough that revealed insight into the genetic basis of obesity. This mini-review aims to review landmark studies on the physiologic system of body weight control. The basic research on the leptin system has broad implications for the genetic control of body weight, thus contributing to solve the global obesity crisis.


Asunto(s)
Peso Corporal/genética , Leptina/genética , Leptina/metabolismo , Obesidad/genética , Animales , Distinciones y Premios , Sistema Endocrino/fisiología , Homeostasis , Humanos , Hipotálamo/fisiología , Vías Nerviosas/fisiología
18.
Trends Cell Biol ; 31(11): 869-872, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34535364

RESUMEN

Meal timing resets trillions of cellular circadian clocks in the body. Recent advances in multiomics demonstrate that clocks in peripheral tissues are differentially reset by feeding rhythm, and modulated by the central clock and the liver clock. This highlights the essential roles of tissue-specific regulation and intercellular signaling in clock synchronization.


Asunto(s)
Relojes Circadianos , Núcleo Supraquiasmático , Ritmo Circadiano/fisiología , Humanos , Hígado , Fotoperiodo , Núcleo Supraquiasmático/fisiología
19.
Front Genet ; 12: 721231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557221

RESUMEN

The circadian clock coordinates physiology, metabolism, and behavior with the 24-h cycles of environmental light. Fundamental mechanisms of how the circadian clock regulates organ physiology and metabolism have been elucidated at a rapid speed in the past two decades. Here we review circadian networks in more than six organ systems associated with complex disease, which cluster around metabolic disorders, and seek to propose critical regulatory molecules controlled by the circadian clock (named clock-controlled checkpoints) in the pathogenesis of complex disease. These include clock-controlled checkpoints such as circadian nuclear receptors in liver and muscle tissues, chemokines and adhesion molecules in the vasculature. Although the progress is encouraging, many gaps in the mechanisms remain unaddressed. Future studies should focus on devising time-dependent strategies for drug delivery and engagement in well-characterized organs such as the liver, and elucidating fundamental circadian biology in so far less characterized organ systems, including the heart, blood, peripheral neurons, and reproductive systems.

20.
STAR Protoc ; 2(3): 100701, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34382024

RESUMEN

Inverted feeding is a paradigm to study synchronization of circadian clocks by feeding rhythm in tissues more directly. Here, we provide a protocol for performing inverted feeding in mice and analyzing circadian rhythmicity in mouse tissues. We describe setting up inverted feeding and performing tissue dissection, followed by RNA extraction and gene expression analysis, and lastly R software-based analysis of circadian rhythmicity. This protocol can be combined with the use of CircaMetDB database for mechanistic studies of inverted feeding. For complete details on the use and execution of this protocol, please refer to Xin et al. (2021).


Asunto(s)
Ritmo Circadiano/fisiología , Biología Computacional/métodos , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Conducta Alimentaria/fisiología , Metabolismo/fisiología , Ratones , Proteínas Circadianas Period/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA