Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.761
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 175(3): 695-708.e13, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30293865

RESUMO

We have uncovered the existence of extracellular vesicle (EV)-mediated signaling between cell types within the adipose tissue (AT) proper. This phenomenon became evident in our attempts at generating an adipocyte-specific knockout of caveolin 1 (cav1) protein. Although we effectively ablated the CAV1 gene in adipocytes, cav1 protein remained abundant. With the use of newly generated mouse models, we show that neighboring endothelial cells (ECs) transfer cav1-containing EVs to adipocytes in vivo, which reciprocate by releasing EVs to ECs. AT-derived EVs contain proteins and lipids capable of modulating cellular signaling pathways. Furthermore, this mechanism facilitates transfer of plasma constituents from ECs to the adipocyte. The transfer event is physiologically regulated by fasting/refeeding and obesity, suggesting EVs participate in the tissue response to changes in the systemic nutrient state. This work offers new insights into the complex signaling mechanisms that exist among adipocytes, stromal vascular cells, and, potentially, distal organs.


Assuntos
Adipócitos/metabolismo , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Jejum/metabolismo , Transdução de Sinais , Animais , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular , Células Cultivadas , Endotélio Vascular/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Cell ; 165(3): 566-79, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27087445

RESUMO

Hepatic glucose release into the circulation is vital for brain function and survival during periods of fasting and is modulated by an array of hormones that precisely regulate plasma glucose levels. We have identified a fasting-induced protein hormone that modulates hepatic glucose release. It is the C-terminal cleavage product of profibrillin, and we name it Asprosin. Asprosin is secreted by white adipose, circulates at nanomolar levels, and is recruited to the liver, where it activates the G protein-cAMP-PKA pathway, resulting in rapid glucose release into the circulation. Humans and mice with insulin resistance show pathologically elevated plasma asprosin, and its loss of function via immunologic or genetic means has a profound glucose- and insulin-lowering effect secondary to reduced hepatic glucose release. Asprosin represents a glucogenic protein hormone, and therapeutically targeting it may be beneficial in type II diabetes and metabolic syndrome.


Assuntos
Jejum/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/metabolismo , Tecido Adiposo Branco/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/administração & dosagem , Ritmo Circadiano , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Jejum/sangue , Feminino , Retardo do Crescimento Fetal/metabolismo , Fibrilina-1 , Glucose/metabolismo , Humanos , Insulina/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteínas dos Microfilamentos/sangue , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Hormônios Peptídicos/sangue , Hormônios Peptídicos/química , Hormônios Peptídicos/genética , Progéria/metabolismo , Proteínas Recombinantes/administração & dosagem , Alinhamento de Sequência
3.
Mol Cell ; 83(7): 1093-1108.e8, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-36863348

RESUMO

The glucagon-PKA signal is generally believed to control hepatic gluconeogenesis via the CREB transcription factor. Here we uncovered a distinct function of this signal in directly stimulating histone phosphorylation for gluconeogenic gene regulation in mice. In the fasting state, CREB recruited activated PKA to regions near gluconeogenic genes, where PKA phosphorylated histone H3 serine 28 (H3S28ph). H3S28ph, recognized by 14-3-3ζ, promoted recruitment of RNA polymerase II and transcriptional stimulation of gluconeogenic genes. In contrast, in the fed state, more PP2A was found near gluconeogenic genes, which counteracted PKA by dephosphorylating H3S28ph and repressing transcription. Importantly, ectopic expression of phosphomimic H3S28 efficiently restored gluconeogenic gene expression when liver PKA or CREB was depleted. These results together highlight a different functional scheme in regulating gluconeogenesis by the glucagon-PKA-CREB-H3S28ph cascade, in which the hormone signal is transmitted to chromatin for rapid and efficient gluconeogenic gene activation.


Assuntos
Glucagon , Gluconeogênese , Animais , Camundongos , Gluconeogênese/genética , Glucagon/metabolismo , Histonas/metabolismo , Fosforilação , Proteínas 14-3-3/metabolismo , Fígado/metabolismo , Jejum/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo
4.
Cell ; 151(7): 1595-607, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260145

RESUMO

Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in ß cells. Here, a mouse genetics approach shows that removal of TCF4 from ß cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease.


Assuntos
Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucose/metabolismo , Fígado/metabolismo , Redes e Vias Metabólicas , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Dieta Hiperlipídica , Jejum/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Knockout , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Ativação Transcricional
5.
Mol Cell ; 76(4): 531-545.e5, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31706703

RESUMO

The glucocorticoid receptor (GR) is a potent metabolic regulator and a major drug target. While GR is known to play integral roles in circadian biology, its rhythmic genomic actions have never been characterized. Here we mapped GR's chromatin occupancy in mouse livers throughout the day and night cycle. We show how GR partitions metabolic processes by time-dependent target gene regulation and controls circulating glucose and triglycerides differentially during feeding and fasting. Highlighting the dominant role GR plays in synchronizing circadian amplitudes, we find that the majority of oscillating genes are bound by and depend on GR. This rhythmic pattern is altered by high-fat diet in a ligand-independent manner. We find that the remodeling of oscillatory gene expression and postprandial GR binding results from a concomitant increase of STAT5 co-occupancy in obese mice. Altogether, our findings highlight GR's fundamental role in the rhythmic orchestration of hepatic metabolism.


Assuntos
Cromatina/metabolismo , Relógios Circadianos , Ritmo Circadiano , Dieta Hiperlipídica , Gorduras na Dieta/metabolismo , Metabolismo Energético , Fígado/metabolismo , Obesidade/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Glicemia/metabolismo , Relógios Circadianos/genética , Ritmo Circadiano/genética , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/sangue , Modelos Animais de Doenças , Metabolismo Energético/genética , Jejum/metabolismo , Regulação da Expressão Gênica , Glucocorticoides/metabolismo , Gluconeogênese , Ligantes , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/genética , PPAR alfa/genética , PPAR alfa/metabolismo , Período Pós-Prandial , Receptores de Glucocorticoides/deficiência , Receptores de Glucocorticoides/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Via Secretória , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Triglicerídeos/sangue
6.
Mol Cell ; 70(3): 531-544.e9, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29727621

RESUMO

While the majority of phosphatidylinositol-4, 5-bisphosphate (PI-4, 5-P2) in mammalian cells is generated by the conversion of phosphatidylinositol-4-phosphate (PI-4-P) to PI-4, 5-P2, a small fraction can be made by phosphorylating phosphatidylinositol-5-phosphate (PI-5-P). The physiological relevance of this second pathway is not clear. Here, we show that deletion of the genes encoding the two most active enzymes in this pathway, Pip4k2a and Pip4k2b, in the liver of mice causes a large enrichment in lipid droplets and in autophagic vesicles during fasting. These changes are due to a defect in the clearance of autophagosomes that halts autophagy and reduces the supply of nutrients salvaged through this pathway. Similar defects in autophagy are seen in nutrient-starved Pip4k2a-/-Pip4k2b-/- mouse embryonic fibroblasts and in C. elegans lacking the PI5P4K ortholog. These results suggest that this alternative pathway for PI-4, 5-P2 synthesis evolved, in part, to enhance the ability of multicellular organisms to survive starvation.


Assuntos
Autofagia/fisiologia , Jejum/metabolismo , Metabolismo dos Lipídeos/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Autofagossomos/metabolismo , Caenorhabditis elegans/metabolismo , Linhagem Celular , Fibroblastos/metabolismo , Células HEK293 , Humanos , Fígado/metabolismo , Camundongos , Fosfatos de Fosfatidilinositol/metabolismo , Transdução de Sinais/fisiologia
7.
Metabolomics ; 20(3): 50, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722393

RESUMO

INTRODUCTION: Analysis of time-resolved postprandial metabolomics data can improve our understanding of the human metabolism by revealing similarities and differences in postprandial responses of individuals. Traditional data analysis methods often rely on data summaries or univariate approaches focusing on one metabolite at a time. OBJECTIVES: Our goal is to provide a comprehensive picture in terms of the changes in the human metabolism in response to a meal challenge test, by revealing static and dynamic markers of phenotypes, i.e., subject stratifications, related clusters of metabolites, and their temporal profiles. METHODS: We analyze Nuclear Magnetic Resonance (NMR) spectroscopy measurements of plasma samples collected during a meal challenge test from 299 individuals from the COPSAC2000 cohort using a Nightingale NMR panel at the fasting and postprandial states (15, 30, 60, 90, 120, 150, 240 min). We investigate the postprandial dynamics of the metabolism as reflected in the dynamic behaviour of the measured metabolites. The data is arranged as a three-way array: subjects by metabolites by time. We analyze the fasting state data to reveal static patterns of subject group differences using principal component analysis (PCA), and fasting state-corrected postprandial data using the CANDECOMP/PARAFAC (CP) tensor factorization to reveal dynamic markers of group differences. RESULTS: Our analysis reveals dynamic markers consisting of certain metabolite groups and their temporal profiles showing differences among males according to their body mass index (BMI) in response to the meal challenge. We also show that certain lipoproteins relate to the group difference differently in the fasting vs. dynamic state. Furthermore, while similar dynamic patterns are observed in males and females, the BMI-related group difference is observed only in males in the dynamic state. CONCLUSION: The CP model is an effective approach to analyze time-resolved postprandial metabolomics data, and provides a compact but a comprehensive summary of the postprandial data revealing replicable and interpretable dynamic markers crucial to advance our understanding of changes in the metabolism in response to a meal challenge.


Assuntos
Metabolômica , Período Pós-Prandial , Humanos , Período Pós-Prandial/fisiologia , Masculino , Feminino , Metabolômica/métodos , Adulto , Jejum/metabolismo , Análise de Componente Principal , Espectroscopia de Ressonância Magnética/métodos , Pessoa de Meia-Idade , Análise de Dados , Metaboloma/fisiologia
8.
Cell ; 136(6): 1056-72, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19303849

RESUMO

Fatty acid synthase (FAS) is a central enzyme in lipogenesis and transcriptionally activated in response to feeding and insulin signaling. The transcription factor USF is required for the activation of FAS transcription, and we show here that USF phosphorylation by DNA-PK, which is dephosphorylated by PP1 in response to feeding, triggers a switch-like mechanism. Under fasting conditions, USF-1 is deacetylated by HDAC9, causing promoter inactivation. In contrast, feeding induces the recruitment of DNA-PK to USF-1 and its phosphorylation, which then allows recruitment of P/CAF, resulting in USF-1 acetylation and FAS promoter activation. DNA break/repair components associated with USF induce transient DNA breaks during FAS activation. In DNA-PK-deficient SCID mice, feeding-induced USF-1 phosphorylation/acetylation, DNA breaks, and FAS activation leading to lipogenesis are impaired, resulting in decreased triglyceride levels. Our study demonstrates that a kinase central to the DNA damage response mediates metabolic gene activation.


Assuntos
Proteína Quinase Ativada por DNA/metabolismo , Insulina/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Ingestão de Alimentos/fisiologia , Jejum/metabolismo , Ácido Graxo Sintases/metabolismo , Histona Desacetilases/metabolismo , Humanos , Camundongos , Camundongos SCID , Fosforilação , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Fatores Estimuladores Upstream/metabolismo
9.
Nucleic Acids Res ; 50(10): 5528-5544, 2022 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-35556130

RESUMO

During fasting, hepatocytes produce glucose in response to hormonal signals. Glucagon and glucocorticoids are principal fasting hormones that cooperate in regulating glucose production via gluconeogenesis. However, how these hormone signals are integrated and interpreted to a biological output is unknown. Here, we use genome-wide profiling of gene expression, enhancer dynamics and transcription factor (TF) binding in primary mouse hepatocytes to uncover the mode of cooperation between glucagon and glucocorticoids. We found that compared to a single treatment with each hormone, a dual treatment directs hepatocytes to a pro-gluconeogenic gene program by synergistically inducing gluconeogenic genes. The cooperative mechanism driving synergistic gene expression is based on 'assisted loading' whereby a glucagon-activated TF (cAMP responsive element binding protein; CREB) leads to enhancer activation which facilitates binding of the glucocorticoid receptor (GR) upon glucocorticoid stimulation. Glucagon does not only activate single enhancers but also activates enhancer clusters, thereby assisting the loading of GR also across enhancer units within the cluster. In summary, we show that cells integrate extracellular signals by an enhancer-specific mechanism: one hormone-activated TF activates enhancers, thereby assisting the loading of a TF stimulated by a second hormone, leading to synergistic gene induction and a tailored transcriptional response to fasting.


Assuntos
Jejum , Glucagon , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Jejum/metabolismo , Glucagon/metabolismo , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Gluconeogênese/genética , Glucose/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Camundongos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
10.
Metab Brain Dis ; 39(4): 635-648, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38429463

RESUMO

Obesity results from an energy imbalance and has been considered an epidemic due to its increasing rates worldwide. It is classified as a low-grade chronic inflammatory disease and has associated comorbidities. Different nutritional strategies are used for the purpose of weight loss, highlighting low-carbohydrate (LC) diets, ketogenic diets, and intermittent fasting (IF). These strategies can lead to metabolic and behavioral changes as they stimulate different biochemical pathways. Therefore, this study evaluated memory, energy metabolism, neuroinflammation, oxidative stress, and antioxidant defense parameters in mice subjected to an LC diet, ketogenic diet (KD), or IF. Eighty male Swiss mice, 60 days old, were divided into 4 groups: control, LC, KD, or IF. Body weight was measured weekly, and food intake every 48 h. After 15 days of nutritional interventions, the animals were subjected to the behavioral object recognition test and subsequently euthanized. Then, visceral fat was removed and weighed, and the brain was isolated for inflammatory and biochemical analysis. We concluded from this study that the LC and KD strategies could damage memory, IF improves the production of adenosine triphosphate (ATP), and the LC, KD, and IF strategies do not lead to neuroinflammatory damage but present damage at the level of oxidative stress.


Assuntos
Dieta Cetogênica , Estresse Oxidativo , Animais , Masculino , Camundongos , Estresse Oxidativo/fisiologia , Transtornos da Memória/metabolismo , Transtornos da Memória/etiologia , Doenças Neuroinflamatórias/metabolismo , Dieta com Restrição de Carboidratos , Jejum/metabolismo , Metabolismo Energético/fisiologia , Encéfalo/metabolismo
11.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34732569

RESUMO

Abnormalities of lipid/lipoprotein and glucose metabolism are hallmarks of hepatic insulin resistance in type 2 diabetes. The former antedate the latter, but the latter become progressively refractory to treatment and contribute to therapeutic failures. It's unclear whether the two processes share a common pathogenesis and what underlies their progressive nature. In this study, we investigated the hypothesis that genes in the lipid/lipoprotein pathway and those in the glucose metabolic pathway are governed by different transcriptional regulatory logics that affect their response to physiologic (fasting/refeeding) as well as pathophysiologic cues (insulin resistance and hyperglycemia). To this end, we obtained genomic and transcriptomic maps of the key insulin-regulated transcription factor, FoxO1, and integrated them with those of CREB, PPAR-α, and glucocorticoid receptor. We found that glucose metabolic genes are primarily regulated by promoter and intergenic enhancers in a fasting-dependent manner, while lipid genes are regulated through fasting-dependent intron enhancers and fasting-independent enhancerless introns. Glucose genes also showed a remarkable transcriptional resiliency (i.e., the ability to compensate following constitutive FoxO1 ablation through an enrichment of active marks at shared PPAR-α/FoxO1 regulatory elements). Unexpectedly, insulin resistance and hyperglycemia were associated with a "spreading" of FoxO1 binding to enhancers and the emergence of unique target sites. We surmise that this unusual pattern correlates with the progressively intractable nature of hepatic insulin resistance. This transcriptional logic provides an integrated model to interpret the combined lipid and glucose abnormalities of type 2 diabetes.


Assuntos
Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Resistência à Insulina , Fígado/metabolismo , Modelos Biológicos , Animais , Jejum/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transcrição Gênica
12.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33723074

RESUMO

Fasting in mammals promotes increases in circulating glucagon and decreases in circulating insulin that stimulate catabolic programs and facilitate a transition from glucose to lipid burning. The second messenger cAMP mediates effects of glucagon on fasting metabolism, in part by promoting the phosphorylation of CREB and the dephosphorylation of the cAMP-regulated transcriptional coactivators (CRTCs) in hepatocytes. In Drosophila, fasting also triggers activation of the single Crtc homolog in neurons, via the PKA-mediated phosphorylation and inhibition of salt-inducible kinases. Crtc mutant flies are more sensitive to starvation and oxidative stress, although the underlying mechanism remains unclear. Here we use RNA sequencing to identify Crtc target genes that are up-regulated in response to starvation. We found that Crtc stimulates a subset of fasting-inducible genes that have conserved CREB binding sites. In keeping with its role in the starvation response, Crtc was found to induce the expression of genes that inhibit insulin secretion (Lst) and insulin signaling (Impl2). In parallel, Crtc also promoted the expression of genes involved in one-carbon (1-C) metabolism. Within the 1-C pathway, Crtc stimulated the expression of enzymes that encode modulators of S-adenosyl-methionine metabolism (Gnmt and Sardh) and purine synthesis (ade2 and AdSl) Collectively, our results point to an important role for the CREB/CRTC pathway in promoting energy balance in the context of nutrient stress.


Assuntos
Proteínas de Drosophila/genética , Metabolismo Energético , Jejum/metabolismo , Insulina/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Animais , Carbono/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Regulação Enzimológica da Expressão Gênica , Ligação Proteica , Estresse Fisiológico , Fatores de Transcrição/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-38432457

RESUMO

Male Japanese quails (Coturnix japonica) have been found to exhibit a three-phase metabolic change when subjected to prolonged fasting, during which basal thermogenesis is significantly reduced. A study had shown that there is a significant difference in the body temperature between male and female Japanese quails. However, whether female Japanese quails also show the same characteristic three-phase metabolic change during prolonged fasting and the underlying thermogenesis mechanisms associated with such changes are still unclear. In this study, female Japanese quails were subjected to prolonged starvation, and the body mass, basal metabolic rate (BMR), body temperature, mass of tissues and organs, body fat content, the state-4 respiration (S4R) and cytochrome c oxidase (CCO) activity in the muscle and liver of these birds were measured to determine the status of metabolic changes triggered by the starvation. In addition, the levels of glucose, triglyceride (TG) and uric acid, and thyroid hormones (T3 and T4) in the serum and the mRNA levels of myostatin (MSTN) and avian uncoupling protein (av-UCP) in the muscle were also measured. The results revealed the existence of a three-phase stage similar to that found in male Japanese quails undergoing prolonged starvation. Fasting resulted in significantly lower body mass, BMR, body temperature, tissues masses and most organs masses, as well as S4R and CCO activity in the muscle and liver. The mRNA level of av-UCP decreased during fasting, while that of MSTN increased but only during Phase I and II and decreased significantly during Phase III. Fasting also significantly lowered the T3 level and the ratio of T3/T4 in the serum. These results indicated that female Japanese quails showed an adaptive response in basal thermogenesis at multiple hierarchical levels, from organismal to biochemical, enzyme and cellular level, gene and endocrine levels and this integrated adjustment could be a part of the adaptation used by female quails to survive long-term fasting.


Assuntos
Coturnix , Codorniz , Feminino , Masculino , Animais , Coturnix/metabolismo , Codorniz/metabolismo , Jejum/metabolismo , Termogênese , RNA Mensageiro/genética
14.
Am J Physiol Endocrinol Metab ; 325(5): E500-E512, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37672249

RESUMO

The ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway are the primary means of degradation in mammalian tissues. We sought to determine the individual contribution of the UPS and autophagy to tissue catabolism during fasting. Mice were overnight fasted for 15 h before regaining food access ("Fed" group, n = 6) or continuing to fast ("Fast" group, n = 7) for 3 h. In addition, to investigate the effects of autophagy on systemic metabolism and tissue degradation, one group of mice was fasted for 18 h and treated with chloroquine ("Fast + CLQ" group, n = 7) and a fourth group of mice was treated with bortezomib ("Fast + Bort" group, n = 7) to assess the contribution of the UPS. Body weight, tissue weight, circulating hormones and metabolites, intracellular signaling pathways, and protein synthesis were investigated. Fasting induced the loss of body weight, liver mass, and white adipose tissue in the Fast and the Fast + CLQ group, whereas the Fast + Bort group maintained tissue and body weight. Fasting reduced glucose and increased ß hydroxybutyrate in the circulation of all mice. Both changes were most profound in the Fast + Bort group compared with the other fasting conditions. Molecular signaling indicated a successful inhibition of hepatic UPS with bortezomib and an upregulation of the PI3K/AKT/mTOR pathway. The latter was further supported by an increase in hepatic protein synthesis with bortezomib. Inhibition of the UPS through bortezomib blocks body weight loss and tissue catabolism during an acute overnight fast in mice. The effects were likely mediated through a combined effect of the drug on biomolecule degradation and synthesis.NEW & NOTEWORTHY Bortezomib treatment prevents tissue and body weight loss during fasting. The loss of proteasome activity with bortezomib exacerbates fasting-induced ketogenesis. During fasting, bortezomib increases AMPK and PI3K/AKT signaling in the liver, which promotes protein synthesis.


Assuntos
Fosfatidilinositol 3-Quinases , Complexo de Endopeptidases do Proteassoma , Camundongos , Animais , Complexo de Endopeptidases do Proteassoma/metabolismo , Bortezomib/farmacologia , Proteólise , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Jejum/metabolismo , Nutrientes , Redução de Peso , Peso Corporal , Autofagia , Mamíferos/metabolismo
15.
Biochem Biophys Res Commun ; 650: 132-136, 2023 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-36796223

RESUMO

Keratins are key structural proteins found in skin and other epithelial tissues. Keratins also protect epithelial cells from damage or stress. Fifty-four human keratins were identified and classified into two families, type I and type II. Accumulating studies showed that keratin expression is highly tissue-specific and used as a diagnostic marker for human diseases. Notably, keratin 79 (KRT79) is type II cytokeratin that was identified as regulator of hair canal morphogenesis and regeneration in skin, but its role in liver remains unclear. KRT79 is undetectable in normal mouse but its expression is significantly increased by the PPARA agonist WY-14643 and fenofibrate, and completely abolished in Ppara-null mice. The Krt79 gene has functional PPARA binding element between exon 1 and exon 2. Hepatic Krt79 is regulated by HNF4A and HER2. Moreover, hepatic KRT79 is also significantly elevated by fasting- and high-fat diet-induced stress, and these increases are completely abolished in Ppara-null mice. These findings suggest that hepatic KRT79 is controlled by PPARA and is highly associated with liver damage. Thus, KRT79 may be considered as a diagnostic marker for human liver diseases.


Assuntos
Hepatopatias , Fígado , Humanos , Camundongos , Animais , Fígado/metabolismo , Queratinas/metabolismo , Hepatopatias/metabolismo , Cabelo/metabolismo , Jejum/metabolismo , Camundongos Knockout
16.
Nat Rev Neurosci ; 19(2): 63-80, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29321682

RESUMO

During evolution, individuals whose brains and bodies functioned well in a fasted state were successful in acquiring food, enabling their survival and reproduction. With fasting and extended exercise, liver glycogen stores are depleted and ketones are produced from adipose-cell-derived fatty acids. This metabolic switch in cellular fuel source is accompanied by cellular and molecular adaptations of neural networks in the brain that enhance their functionality and bolster their resistance to stress, injury and disease. Here, we consider how intermittent metabolic switching, repeating cycles of a metabolic challenge that induces ketosis (fasting and/or exercise) followed by a recovery period (eating, resting and sleeping), may optimize brain function and resilience throughout the lifespan, with a focus on the neuronal circuits involved in cognition and mood. Such metabolic switching impacts multiple signalling pathways that promote neuroplasticity and resistance of the brain to injury and disease.


Assuntos
Encéfalo/fisiologia , Jejum/metabolismo , Plasticidade Neuronal/fisiologia , Animais , Humanos
17.
FASEB J ; 36(2): e22152, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35061305

RESUMO

Catabolic conditions, such as starvation, inactivity, and cancer cachexia, induce Forkhead box O (FOXO) transcription factor(s) expression and severe muscle atrophy via the induction of ubiquitin-proteasome system-mediated muscle proteolysis, resulting in frailty and poor quality of life. Although FOXOs are clearly essential for the induction of muscle atrophy, it is unclear whether there are other factors involved in the FOXO-mediated transcriptional regulation. As such, we identified FOXO-CCAAT/enhancer-binding protein δ (C/EBPδ) signaling pathway as a novel proteolytic pathway. By comparing the gene expression profiles of FOXO1-transgenic (gain-of-function model) and FOXO1,3a,4-/- (loss-of-function model) mice, we identified several novel FOXO1-target genes in skeletal muscle including Redd1, Sestrin1, Castor2, Chac1, Depp1, Lat3, as well as C/EBPδ. During starvation, C/EBPδ abundance was increased in a FOXOs-dependent manner. Notably, knockdown of C/EBPδ prevented the induction of the ubiquitin-proteasome system and decrease of myofibers in FOXO1-activated myotubes. Conversely, C/EBPδ overexpression in primary myotubes induced myotube atrophy. Furthermore, we demonstrated that FOXO1 enhances the promoter activity of target genes in cooperation with C/EBPδ and ATF4. This research comprehensively identifies novel FOXO1 target genes in skeletal muscle and clarifies the pathophysiological role of FOXO1, a master regulator of skeletal muscle atrophy.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Jejum/metabolismo , Proteína Forkhead Box O1/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Transcrição Gênica/fisiologia , Animais , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Transdução de Sinais/fisiologia , Ubiquitina/metabolismo
18.
Nature ; 543(7645): 433-437, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28199315

RESUMO

Mechanistic target of rapamycin complex 1 (TORC1) integrates nutrient signals to control cell growth and organismal homeostasis across eukaryotes. The evolutionarily conserved GATOR complex regulates mTORC1 signalling through Rag GTPases, and GATOR1 displays GTPase activating protein (GAP) activity for RAGA and RAGB (RAGA/B) and GATOR2 has been proposed to be an inhibitor of GATOR1. Furthermore, the metazoan-specific SESN proteins function as guanine nucleotide dissociation inhibitors (GDIs) for RAGA/B, and interact with GATOR2 with unknown effects. Here we show that SZT2 (seizure threshold 2), a metazoan-specific protein mutated in epilepsy, recruits a fraction of mammalian GATOR1 and GATOR2 to form a SZT2-orchestrated GATOR (SOG) complex with an essential role in GATOR- and SESN-dependent nutrient sensing and mTORC1 regulation. The interaction of SZT2 with GATOR1 and GATOR2 was synergistic, and an intact SOG complex was required for its localization at the lysosome. SZT2 deficiency resulted in constitutive mTORC1 signalling in cells under nutrient-deprived conditions and neonatal lethality in mice, which was associated with failure to inactivate mTORC1 during fasting. Hyperactivation of mTORC1 in SZT2-deficient cells could be partially corrected by overexpression of the GATOR1 component DEPDC5, and by the lysosome-targeted GATOR2 component WDR59 or lysosome-targeted SESN2. These findings demonstrate that SZT2 has a central role in dictating GATOR-dependent nutrient sensing by promoting lysosomal localization of SOG, and reveal an unexpected function of lysosome-located GATOR2 in suppressing mTORC1 signalling through SESN recruitment.


Assuntos
Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Jejum/metabolismo , Feminino , Alimentos , Privação de Alimentos , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/química , Proteínas do Tecido Nervoso/deficiência , Proteínas Nucleares/metabolismo , Peroxidases , Proteínas Repressoras/metabolismo
19.
Nature ; 551(7678): 115-118, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29045397

RESUMO

Mammalian tissues are fuelled by circulating nutrients, including glucose, amino acids, and various intermediary metabolites. Under aerobic conditions, glucose is generally assumed to be burned fully by tissues via the tricarboxylic acid cycle (TCA cycle) to carbon dioxide. Alternatively, glucose can be catabolized anaerobically via glycolysis to lactate, which is itself also a potential nutrient for tissues and tumours. The quantitative relevance of circulating lactate or other metabolic intermediates as fuels remains unclear. Here we systematically examine the fluxes of circulating metabolites in mice, and find that lactate can be a primary source of carbon for the TCA cycle and thus of energy. Intravenous infusions of 13C-labelled nutrients reveal that, on a molar basis, the circulatory turnover flux of lactate is the highest of all metabolites and exceeds that of glucose by 1.1-fold in fed mice and 2.5-fold in fasting mice; lactate is made primarily from glucose but also from other sources. In both fed and fasted mice, 13C-lactate extensively labels TCA cycle intermediates in all tissues. Quantitative analysis reveals that during the fasted state, the contribution of glucose to tissue TCA metabolism is primarily indirect (via circulating lactate) in all tissues except the brain. In genetically engineered lung and pancreatic cancer tumours in fasted mice, the contribution of circulating lactate to TCA cycle intermediates exceeds that of glucose, with glutamine making a larger contribution than lactate in pancreatic cancer. Thus, glycolysis and the TCA cycle are uncoupled at the level of lactate, which is a primary circulating TCA substrate in most tissues and tumours.


Assuntos
Ciclo do Ácido Cítrico , Glucose/metabolismo , Ácido Láctico/sangue , Ácido Láctico/metabolismo , Animais , Glicemia/metabolismo , Encéfalo/metabolismo , Carbono/sangue , Carbono/metabolismo , Jejum/sangue , Jejum/metabolismo , Glutamina/sangue , Glutamina/metabolismo , Glicólise , Camundongos , Músculos/metabolismo , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/metabolismo
20.
BMC Geriatr ; 23(1): 217, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37020298

RESUMO

BACKGROUND: During biological aging, significant metabolic dysregulation in the central nervous system may lead to cognitive decline and neurodegeneration. However, the metabolomics of the aging process in cerebrospinal fluid (CSF) has not been thoroughly explored. METHODS: In this cohort study of CSF metabolomics using liquid chromatography-mass spectrometry (LC-MS), fasting CSF samples collected from 92 cognitively unimpaired adults aged 20-87 years without obesity or diabetes were analyzed. RESULTS: We identified 37 metabolites in these CSF samples with significant positive correlations with aging, including cysteine, pantothenic acid, 5-hydroxyindoleacetic acid (5-HIAA), aspartic acid, and glutamate; and two metabolites with negative correlations, asparagine and glycerophosphocholine. The combined alterations of asparagine, cysteine, glycerophosphocholine, pantothenic acid, sucrose, and 5-HIAA showed a superior correlation with aging (AUC = 0.982). These age-correlated changes in CSF metabolites might reflect blood-brain barrier breakdown, neuroinflammation, and mitochondrial dysfunction in the aging brain. We also found sex differences in CSF metabolites with higher levels of taurine and 5-HIAA in women using propensity-matched comparison. CONCLUSIONS: Our LC-MS metabolomics of the aging process in a Taiwanese population revealed several significantly altered CSF metabolites during aging and between the sexes. These metabolic alterations in CSF might provide clues for healthy brain aging and deserve further exploration.


Assuntos
Envelhecimento , Cromatografia Líquida , Cisteína , Metaboloma , Espectrometria de Massas em Tandem , Feminino , Humanos , Masculino , Envelhecimento/líquido cefalorraquidiano , Envelhecimento/metabolismo , Asparagina/líquido cefalorraquidiano , Cromatografia Líquida/métodos , Estudos de Coortes , Cisteína/líquido cefalorraquidiano , Ácido Hidroxi-Indolacético/líquido cefalorraquidiano , Ácido Pantotênico/líquido cefalorraquidiano , Espectrometria de Massas em Tandem/métodos , Voluntários Saudáveis , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Cognição/fisiologia , Jejum/líquido cefalorraquidiano , Jejum/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA