Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 757
Filtrar
1.
Immunity ; 55(2): 210-223, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35139351

RESUMO

Nutrition affects all physiological processes including those linked to the development and function of our immune system. Here, we discuss recent evidence and emerging concepts supporting the idea that our newfound relationship with nutrition in industrialized countries has fundamentally altered the way in which our immune system is wired. This will be examined through the lens of studies showing that mild or transient reductions in dietary intake can enhance protective immunity while also limiting aberrant inflammatory responses. We will further discuss how trade-offs and priorities begin to emerge in the context of severe nutritional stress. In those settings, specific immunological functions are heightened to re-enforce processes and tissue sites most critical to survival. Altogether, these examples will emphasize the profound influence nutrition has over the immune system and highlight how a mechanistic exploration of this cross talk could ultimately lead to the design of novel therapeutic approaches that prevent and treat disease.


Assuntos
Dietoterapia , Imunidade , Envelhecimento/imunologia , Restrição Calórica , Humanos , Inflamação , Corpos Cetônicos/biossíntese , Corpos Cetônicos/imunologia , Desnutrição/imunologia , Microbiota/imunologia , Fenômenos Fisiológicos da Nutrição/imunologia
2.
Nat Metab ; 3(2): 196-210, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33619377

RESUMO

Ketone bodies are generated in the liver and allow for the maintenance of systemic caloric and energy homeostasis during fasting and caloric restriction. It has previously been demonstrated that neonatal ketogenesis is activated independently of starvation. However, the role of ketogenesis during the perinatal period remains unclear. Here, we show that neonatal ketogenesis plays a protective role in mitochondrial function. We generated a mouse model of insufficient ketogenesis by disrupting the rate-limiting hydroxymethylglutaryl-CoA synthase 2 enzyme gene (Hmgcs2). Hmgcs2 knockout (KO) neonates develop microvesicular steatosis within a few days of birth. Electron microscopic analysis and metabolite profiling indicate a restricted energy production capacity and accumulation of acetyl-CoA in Hmgcs2 KO mice. Furthermore, acetylome analysis of Hmgcs2 KO cells revealed enhanced acetylation of mitochondrial proteins. These findings suggest that neonatal ketogenesis protects the energy-producing capacity of mitochondria by preventing the hyperacetylation of mitochondrial proteins.


Assuntos
Metabolismo Energético/fisiologia , Corpos Cetônicos/biossíntese , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Acetilação , Animais , Animais Recém-Nascidos , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Microvasos/fisiologia , Consumo de Oxigênio
3.
J Inherit Metab Dis ; 44(1): 42-53, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32654164

RESUMO

Metabolic epilepsies arise in the context of rare inborn errors of metabolism (IEM), notably glucose transporter type 1 deficiency syndrome, succinic semialdehyde dehydrogenase deficiency, pyruvate dehydrogenase complex deficiency, nonketotic hyperglycinemia, and mitochondrial cytopathies. A common feature of these disorders is impaired bioenergetics, which through incompletely defined mechanisms result in a wide spectrum of neurological symptoms, such as epileptic seizures, developmental delay, and movement disorders. The ketogenic diet (KD) has been successfully utilized to treat such conditions to varying degrees. While the mechanisms underlying the clinical efficacy of the KD in IEM remain unclear, it is likely that the proposed heterogeneous targets influenced by the KD work in concert to rectify or ameliorate the downstream negative consequences of genetic mutations affecting key metabolic enzymes and substrates-such as oxidative stress and cell death. These beneficial effects can be broadly grouped into restoration of impaired bioenergetics and synaptic dysfunction, improved redox homeostasis, anti-inflammatory, and epigenetic activity. Hence, it is conceivable that the KD might prove useful in other metabolic disorders that present with epileptic seizures. At the same time, however, there are notable contraindications to KD use, such as fatty acid oxidation disorders. Clearly, more research is needed to better characterize those metabolic epilepsies that would be amenable to ketogenic therapies, both experimentally and clinically. In the end, the expanded knowledge base will be critical to designing metabolism-based treatments that can afford greater clinical efficacy and tolerability compared to current KD approaches, and improved long-term outcomes for patients.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/dietoterapia , Dieta Cetogênica , Epilepsia/dietoterapia , Corpos Cetônicos/biossíntese , Animais , Contraindicações , Metabolismo Energético , Humanos , Corpos Cetônicos/uso terapêutico , Resultado do Tratamento
4.
J Alzheimers Dis ; 78(3): 887-904, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33074226

RESUMO

Alzheimer's disease (AD) is an irreversible neurodegenerative disease characterized by brain function disorder and chronic cognitive function impairment. The onset of AD is complex and is mostly attributed to interactions between genetic factors and environmental factors. Lifestyle, dietary habits, and food consumption are likely to play indispensable functions in aged-related neurodegenerative diseases in elderly people. An increasing number of epidemiological studies have linked dietary fatty acid factors to AD, raising the point of view that fatty acid metabolism plays an important role in AD initiation and progression as well as in other central nervous system disorders. In this paper, we review the effects of the consumption of various dietary fatty acids on AD onset and progression and discuss the detrimental and beneficial effects of some typical fatty acids derived from dietary patterns on the pathology of AD. We outline these recent advances, and we recommend that healthy dietary lifestyles may contribute to preventing the occurrence and decreasing the pathology of AD.


Assuntos
Doença de Alzheimer/metabolismo , Gorduras na Dieta/metabolismo , Ácidos Graxos/metabolismo , Doença de Alzheimer/epidemiologia , Dieta/estatística & dados numéricos , Dieta Mediterrânea/estatística & dados numéricos , Progressão da Doença , Ácidos Graxos Monoinsaturados , Ácidos Graxos Insaturados , Humanos , Corpos Cetônicos/biossíntese , Metabolismo dos Lipídeos , Lipoproteínas/metabolismo , Oxirredução , Fatores de Proteção , Fatores de Risco
5.
Crit Care ; 24(1): 536, 2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32867803

RESUMO

BACKGROUND: In critically ill children, omitting early use of parenteral nutrition (late-PN versus early-PN) reduced infections, accelerated weaning from mechanical ventilation, and shortened PICU stay. We hypothesized that fasting-induced ketogenesis mediates these benefits. METHODS: In a secondary analysis of the PEPaNIC RCT (N = 1440), the impact of late-PN versus early-PN on plasma 3-hydroxybutyrate (3HB), and on blood glucose, plasma insulin, and glucagon as key ketogenesis regulators, was determined for 96 matched patients staying ≥ 5 days in PICU, and the day of maximal 3HB-effect, if any, was identified. Subsequently, in the total study population, plasma 3HB and late-PN-affected ketogenesis regulators were measured on that average day of maximal 3HB effect. Multivariable Cox proportional hazard and logistic regression analyses were performed adjusting for randomization and baseline risk factors. Whether any potential mediator role for 3HB was direct or indirect was assessed by further adjusting for ketogenesis regulators. RESULTS: In the matched cohort (n = 96), late-PN versus early-PN increased plasma 3HB throughout PICU days 1-5 (P < 0.0001), maximally on PICU day 2. Also, blood glucose (P < 0.001) and plasma insulin (P < 0.0001), but not glucagon, were affected. In the total cohort (n = 1142 with available plasma), late-PN increased plasma 3HB on PICU day 2 (day 1 for shorter stayers) from (median [IQR]) 0.04 [0.04-0.04] mmol/L to 0.75 [0.04-2.03] mmol/L (P < 0.0001). The 3HB effect of late-PN statistically explained its impact on weaning from mechanical ventilation (P = 0.0002) and on time to live PICU discharge (P = 0.004). Further adjustment for regulators of ketogenesis did not alter these findings. CONCLUSION: Withholding early-PN in critically ill children significantly increased plasma 3HB, a direct effect that statistically mediated an important part of its outcome benefit.


Assuntos
Corpos Cetônicos/biossíntese , Nutrição Parenteral , Suspensão de Tratamento , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Unidades de Terapia Intensiva Pediátrica , Masculino , Resultado do Tratamento
6.
Nat Metab ; 2(1): 50-61, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-32694683

RESUMO

Ketone bodies are essential alternative fuels that allow humans to survive periods of glucose scarcity induced by starvation and prolonged exercise. A widely used ketogenic diet (KD), which is extremely high in fat with very low carbohydrates, drives the host into using ß-hydroxybutyrate for the production of ATP and lowers NLRP3-mediated inflammation. However, the extremely high fat composition of KD raises the question of how ketogenesis affects adipose tissue to control inflammation and energy homeostasis. Here, by using single-cell RNA sequencing of adipose-tissue-resident immune cells, we show that KD expands metabolically protective γδ T cells that restrain inflammation. Notably, long-term ad libitum KD feeding in mice causes obesity, impairs metabolic health and depletes the adipose-resident γδ T cells. In addition, mice lacking γδ T cells have impaired glucose homeostasis. Our results suggest that γδ T cells are mediators of protective immunometabolic responses that link fatty acid-driven fuel use to reduced adipose tissue inflammation.


Assuntos
Gordura Intra-Abdominal/metabolismo , Corpos Cetônicos/biossíntese , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Animais , Glicemia/metabolismo , Dieta Cetogênica , Homeostase , Inflamação/metabolismo , Gordura Intra-Abdominal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Linfócitos T/imunologia
7.
Gut ; 69(8): 1423-1431, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31753852

RESUMO

OBJECTIVE: Food intake normally stimulates release of satiety and insulin-stimulating intestinal hormones, such as glucagon-like peptide (GLP)-1. This response is blunted in obese insulin resistant subjects, but is rapidly restored following Roux-en-Y gastric bypass (RYGB) surgery. We hypothesised this to be a result of the metabolic changes taking place in the small intestinal mucosa following the anatomical rearrangement after RYGB surgery, and aimed at identifying such mechanisms. DESIGN: Jejunal mucosa biopsies from patients undergoing RYGB surgery were retrieved before and after very-low calorie diet, at time of surgery and 6 months postoperatively. Samples were analysed by global protein expression analysis and Western blotting. Biological functionality of these findings was explored in mice and enteroendocrine cells (EECs) primary mouse jejunal cell cultures. RESULTS: The most prominent change found after RYGB was decreased jejunal expression of the rate-limiting ketogenic enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mHMGCS), corroborated by decreased ketone body levels. In mice, prolonged high-fat feeding induced the expression of mHMGCS and functional ketogenesis in jejunum. The effect of ketone bodies on gut peptide secretion in EECs showed a ∼40% inhibition of GLP-1 release compared with baseline. CONCLUSION: Intestinal ketogenesis is induced by high-fat diet and inhibited by RYGB surgery. In cell culture, ketone bodies inhibited GLP-1 release from EECs. Thus, we suggest that this may be a mechanism by which RYGB can remove the inhibitory effect of ketone bodies on EECs, thereby restituting the responsiveness of EECs resulting in increased meal-stimulated levels of GLP-1 after surgery.


Assuntos
Restrição Calórica , Células Enteroendócrinas/metabolismo , Derivação Gástrica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Corpos Cetônicos/biossíntese , Ácido 3-Hidroxibutírico/sangue , Ácido 3-Hidroxibutírico/farmacologia , Anastomose em-Y de Roux , Animais , Células Cultivadas , Gorduras na Dieta/administração & dosagem , Emulsões/farmacologia , Emulsões Gordurosas Intravenosas/farmacologia , Feminino , Peptídeo 1 Semelhante ao Glucagon/antagonistas & inibidores , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/metabolismo , Cetonas/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fosfolipídeos/farmacologia , Período Pós-Operatório , Período Pré-Operatório , Cultura Primária de Células , Óleo de Soja/farmacologia
8.
JCI Insight ; 52019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31012869

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent, and potentially morbid, disease that affects one-third of the U.S. population. Normal liver safely accommodates lipid excess during fasting or carbohydrate restriction by increasing their oxidation to acetyl-CoA and ketones, yet lipid excess during NAFLD leads to hyperglycemia and, in some, steatohepatitis. To examine potential mechanisms, flux through pathways of hepatic oxidative metabolism and gluconeogenesis were studied using five simultaneous stable isotope tracers in ketotic (24-hour fast) individuals with a wide range of hepatic triglyceride contents (0-52%). Ketogenesis was progressively impaired as hepatic steatosis and glycemia worsened. Conversely, the alternative pathway for acetyl-CoA metabolism, oxidation in the tricarboxylic (TCA) cycle, was upregulated in NAFLD as ketone production diminished and positively correlated with rates of gluconeogenesis and plasma glucose concentrations. Increased respiration and energy generation that occurred in liver when ß-oxidation and TCA cycle activity were coupled may explain these findings, inasmuch as oxygen consumption was higher during fatty liver and highly correlated with gluconeogenesis. These findings demonstrate that increased glucose production and hyperglycemia in NAFLD is not a consequence of acetyl-CoA production per se, but how acetyl-CoA is further metabolized in liver.


Assuntos
Acetilcoenzima A/metabolismo , Hiperglicemia/metabolismo , Corpos Cetônicos/biossíntese , Cetose/complicações , Hepatopatia Gordurosa não Alcoólica/complicações , Adulto , Glicemia/análise , Ciclo do Ácido Cítrico , Metabolismo Energético , Jejum/fisiologia , Feminino , Gluconeogênese , Técnica Clamp de Glucose , Humanos , Hiperglicemia/sangue , Hiperglicemia/etiologia , Corpos Cetônicos/análise , Cetose/sangue , Cetose/metabolismo , Cetose/fisiopatologia , Fígado/diagnóstico por imagem , Fígado/metabolismo , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Triglicerídeos/análise , Triglicerídeos/metabolismo
9.
In Vitro Cell Dev Biol Anim ; 55(5): 368-375, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31025252

RESUMO

Ketosis is a condition where ketone bodies are produced as an alternative energy source, due to insufficient glucose for energy production so that the body switches from carbohydrate metabolism to mostly fat metabolism. In this study, we examined the anti-ketosis effects of silibinin, a major active component of silymarin. We induced ketosis in FL83B mouse hepatocytes in vitro by culturing in low glucose media and compared results to hepatocytes maintained in high-glucose conditions. We quantified ß-hydroxybutyrate (BHB) levels with a colorimetric assay. In low-glucose conditions, silibinin reduced the amount of BHB produced, compared to high-glucose conditions; thus, silibinin exhibited an anti-ketotic effect. Ketone body formation during beta oxidation is mediated by 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2). The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) regulates the transcription of HMGCS2, and plays a vital role in BHB levels. We showed that silibinin inhibited the expression of HMGCS2 and NF-kB at transcriptional and translational levels. Silibinin also inhibited the nuclear translocation of NF-kB and its DNA binding activity. To elucidate the relationship between HMGCS2 and NF-kB, we tested inhibited and over-expressed NF-kB. We found that NF-kB acted as a positive regulator for HMGCS2 under ketosis treatment conditions.


Assuntos
Hidroximetilglutaril-CoA Sintase/genética , Cetose/tratamento farmacológico , NF-kappa B/genética , Silibina/farmacologia , Ácido 3-Hidroxibutírico/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colorimetria , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Hepatócitos/efeitos dos fármacos , Humanos , Corpos Cetônicos/biossíntese , Corpos Cetônicos/metabolismo , Cetose/genética , Cetose/metabolismo , Cetose/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Transdução de Sinais/efeitos dos fármacos , Silimarina/química , Silimarina/farmacologia
10.
Am J Physiol Gastrointest Liver Physiol ; 316(5): G623-G631, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30767679

RESUMO

Ketosis is a metabolic adaptation to fasting, nonalcoholic fatty liver disease (NAFLD), and prolonged exercise. ß-OH butyrate acts as a transcriptional regulator and at G protein-coupled receptors to modulate cellular signaling pathways in a hormone-like manner. While physiological ketosis is often adaptive, chronic hyperketonemia may contribute to the metabolic dysfunction of NAFLD. To understand how ß-OH butyrate signaling affects hepatic metabolism, we compared the hepatic fasting response in control and 3-hydroxy-3-methylglutaryl-CoA synthase II (HMGCS2) knockdown mice that are unable to elevate ß-OH butyrate production. To establish that rescue of ketone metabolic/endocrine signaling would restore the normal hepatic fasting response, we gave intraperitoneal injections of ß-OH butyrate (5.7 mmol/kg) to HMGCS2 knockdown and control mice every 2 h for the final 9 h of a 16-h fast. In hypoketonemic, HMGCS2 knockdown mice, fasting more robustly increased mRNA expression of uncoupling protein 2 (UCP2), a protein critical for supporting fatty acid oxidation and ketogenesis. In turn, exogenous ß-OH butyrate administration to HMGCS2 knockdown mice decreased fasting UCP2 mRNA expression to that observed in control mice. Also supporting feedback at the transcriptional level, ß-OH butyrate lowered the fasting-induced expression of HMGCS2 mRNA in control mice. ß-OH butyrate also regulates the glycemic response to fasting. The fast-induced fall in serum glucose was absent in HMGCS2 knockdown mice but was restored by ß-OH butyrate administration. These data propose that endogenous ß-OH butyrate signaling transcriptionally regulates hepatic fatty acid oxidation and ketogenesis, while modulating glucose tolerance. NEW & NOTEWORTHY Ketogenesis regulates whole body glucose metabolism and ß-OH butyrate produced by the liver feeds back to inhibit hepatic ß-oxidation and ketogenesis during fasting.


Assuntos
Jejum/fisiologia , Ácidos Graxos/metabolismo , Corpos Cetônicos/biossíntese , Cetonas/metabolismo , Fígado/metabolismo , Adaptação Fisiológica , Animais , Glicemia/metabolismo , Butiratos/metabolismo , Regulação da Expressão Gênica , Hidroximetilglutaril-CoA Sintase/metabolismo , Cetose/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Transdução de Sinais , Proteína Desacopladora 2/metabolismo
11.
Postgrad Med ; 131(2): 82-88, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30757937

RESUMO

Sodium-glucose co-transporter 2 (SGLT2) inhibitors inhibit glucose re-absorption in the proximal renal tubules. Two trials have shown significant reductions of cardiovascular (CV) events with empagliflozin and canagliflozin, which could not be attributed solely to their antidiabetic effects. The aim of the review is the critical presentation of suggested mechanisms/hypotheses for the SGLT2 inhibitors' cardioprotection. The search of the literature revealed many possible cardioprotective mechanisms, because SGLT2 inhibitors (i) increase natriuresis and act as diuretics with unique properties leading to a reduction in preload and myocardial stretch (the diuretic hypothesis); (ii) decrease blood pressure and afterload (the blood pressure lowering hypothesis), (iii) favor the production of ketones, which can act as a 'superfuel' in the cardiac and renal tissue (the 'thrifty substrate' hypothesis), (iv) improve many metabolic variables (the metabolic effects hypothesis), (v) exert many anti-inflammatory effects (the anti-inflammatory effects hypothesis), (vi) can act through the angiotensin II type II receptors in the context of simultaneous renin-angiotensin-aldosterone-system (RAAS) blockade leading to vasodilation and positive inotropic effects (the RAAS hypothesis), (vii) directly decrease the activity of the upregulated in heart failure Na+-H+ exchanger in myocardial cells leading to restoration of mitochondrial calcium handling in cardiomyocytes (the sodium hypothesis). Additionally, some SGLT2 inhibitors exhibit also SGLT1 inhibitory action possibly resulting in an attenuation of oxidative stress in ischemic myocardium (the SGLT1 inhibition hypothesis). Thus, many mechanisms have been suggested (and possibly act cumulatively) for the cardioprotective effects of SGLT2 inhibitors.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Coração/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Natriurese/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Vasodilatação/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Cálcio/metabolismo , Canagliflozina/farmacologia , Cardiotônicos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Glucosídeos/farmacologia , Humanos , Inflamação , Corpos Cetônicos/biossíntese , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Tipo 2 de Angiotensina/efeitos dos fármacos
12.
Cell Metab ; 29(1): 91-102.e5, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30318340

RESUMO

The conversion of lipolysis-derived fatty acids into ketone bodies (ketogenesis) is a crucial metabolic adaptation to prolonged periods of food scarcity. The process occurs primarily in liver mitochondria and is initiated by fatty-acid-mediated stimulation of the ligand-operated transcription factor, peroxisome proliferator-activated receptor-α (PPAR-α). Here, we present evidence that mast cells contribute to the control of fasting-induced ketogenesis via a paracrine mechanism that involves secretion of histamine into the hepatic portal circulation, stimulation of liver H1 receptors, and local biosynthesis of the high-affinity PPAR-α agonist, oleoylethanolamide (OEA). Genetic or pharmacological interventions that disable any one of these events, including mast cell elimination, deletion of histamine- or OEA-synthesizing enzymes, and H1 blockade, blunt ketogenesis without affecting lipolysis. The results reveal an unexpected role for mast cells in the regulation of systemic fatty-acid homeostasis, and suggest that OEA may act in concert with lipolysis-derived fatty acids to activate liver PPAR-α and promote ketogenesis.


Assuntos
Endocanabinoides/metabolismo , Histamina/fisiologia , Corpos Cetônicos/biossíntese , Fígado/metabolismo , Mastócitos/metabolismo , Ácidos Oleicos/metabolismo , PPAR alfa/metabolismo , Animais , Células Hep G2 , Humanos , Fígado/citologia , Masculino , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Histamínicos H1/metabolismo
13.
J Biol Chem ; 294(5): 1579-1589, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30530497

RESUMO

Sirt6 is an NADH (NAD+)-dependent deacetylase with a critical role in hepatic lipid metabolism. Ketogenesis is controlled by a signaling network of hepatic lipid metabolism. However, how Sirt6 functions in ketogenesis remains unclear. Here, we demonstrated that Sirt6 functions as a mediator of ketogenesis in response to a fasting and ketogenic diet (KD). The KD-fed hepatocyte-specific Sirt6 deficiency (HKO) mice exhibited impaired ketogenesis, which was due to enhanced Fsp27 (fat-specific induction of protein 27), a protein known to regulate lipid metabolism. In contrast, overexpression of Sirt6 in mouse primary hepatocytes promoted ketogenesis. Mechanistically, Sirt6 repressed Fsp27ß expression by interacting with Crebh (cAMP response element-binding protein H) and preventing its recruitment to the Fsp27ß gene promoter. The KD-fed HKO mice also showed exacerbated hepatic steatosis and inflammation. Finally, Fsp27 silencing rescued hypoketonemia and other metabolic phenotypes in KD-fed HKO mice. Our data suggest that the Sirt6-Crebh-Fsp27 axis is pivotal for hepatic lipid metabolism and inflammation. Sirt6 may be a pharmacological target to remedy metabolic diseases.


Assuntos
Hepatócitos/metabolismo , Corpos Cetônicos/biossíntese , Proteínas/metabolismo , Sirtuínas/fisiologia , Animais , Hepatócitos/citologia , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
J Am Heart Assoc ; 7(20): e010378, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30371266

RESUMO

Background The molecular mechanisms mediating postnatal loss of cardiac regeneration in mammals are not fully understood. We aimed to provide an integrated resource of mRNA , protein, and metabolite changes in the neonatal heart for identification of metabolism-related mechanisms associated with cardiac regeneration. Methods and Results Mouse ventricular tissue samples taken on postnatal day 1 (P01), P04, P09, and P23 were analyzed with RNA sequencing and global proteomics and metabolomics. Gene ontology analysis, KEGG pathway analysis, and fuzzy c-means clustering were used to identify up- or downregulated biological processes and metabolic pathways on all 3 levels, and Ingenuity pathway analysis (Qiagen) was used to identify upstream regulators. Differential expression was observed for 8547 mRNA s and for 1199 of 2285 quantified proteins. Furthermore, 151 metabolites with significant changes were identified. Differentially regulated metabolic pathways include branched chain amino acid degradation (upregulated at P23), fatty acid metabolism (upregulated at P04 and P09; downregulated at P23) as well as the HMGCS ( HMG -CoA [hydroxymethylglutaryl-coenzyme A] synthase)-mediated mevalonate pathway and ketogenesis (transiently activated). Pharmacological inhibition of HMGCS in primary neonatal cardiomyocytes reduced the percentage of BrdU-positive cardiomyocytes, providing evidence that the mevalonate and ketogenesis routes may participate in regulating the cardiomyocyte cell cycle. Conclusions This study is the first systems-level resource combining data from genomewide transcriptomics with global quantitative proteomics and untargeted metabolomics analyses in the mouse heart throughout the early postnatal period. These integrated data of molecular changes associated with the loss of cardiac regeneration may open up new possibilities for the development of regenerative therapies.


Assuntos
Coração/crescimento & desenvolvimento , Camundongos/crescimento & desenvolvimento , Aminoácidos de Cadeia Ramificada/metabolismo , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Ácidos Graxos/metabolismo , Expressão Gênica/fisiologia , Coração/embriologia , Ventrículos do Coração , Corpos Cetônicos/biossíntese , Metabolômica , Ácido Mevalônico/metabolismo , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/fisiologia , Transcriptoma/fisiologia
15.
Elife ; 72018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30322448

RESUMO

Apart from glucose, fatty acid-derived ketone bodies provide metabolic energy for the brain during fasting and neonatal development. We investigated the evolution of HMGCS2, the key enzyme required for ketone body biosynthesis (ketogenesis). Unexpectedly, we found that three mammalian lineages, comprising cetaceans (dolphins and whales), elephants and mastodons, and Old World fruit bats have lost this gene. Remarkably, many of these species have exceptionally large brains and signs of intelligent behavior. While fruit bats are sensitive to starvation, cetaceans and elephants can still withstand periods of fasting. This suggests that alternative strategies to fuel large brains during fasting evolved repeatedly and reveals flexibility in mammalian energy metabolism. Furthermore, we show that HMGCS2 loss preceded brain size expansion in toothed whales and elephants. Thus, while ketogenesis was likely important for brain size expansion in modern humans, ketogenesis is not a universal precondition for the evolution of large mammalian brains.


Assuntos
Evolução Biológica , Encéfalo/anatomia & histologia , Hidroximetilglutaril-CoA Sintase/genética , Corpos Cetônicos/biossíntese , Mamíferos/anatomia & histologia , Animais , Sequência de Bases , Humanos , Tamanho do Órgão , Regiões Promotoras Genéticas/genética , Deleção de Sequência
16.
Indian J Pediatr ; 85(11): 1000-1005, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30242606

RESUMO

About one-third of childhood epilepsy ultimately becomes drug resistant epilepsy. Only about one-third of drug resistant epilepsy is amenable for epilepsy surgery. Epilepsy surgery and vagal nerve stimulation is still beyond the reach of huge proportion of children with pharmacoresistant epilepsy. Ketogenic diet (KD) has been in use for almost a century now all over the world for drug resistant epilepsy, although in between there was a decline in its popularity with advent of newer antiepileptic drugs like valproate, phenytoin and carbamazepine. Again from 1990s there was resurgence of interest in KD for pharmacoresistant epilepsy and in the last two decades several randomized controlled trials and systemic reviews have proved its efficacy beyond any suspicion. Ketogenic diet is a high fat low carbohydrate and low protein diet, which has been found to reduce epileptogenesis in body most probably by production of ketone bodies. Modified Atkin's Diet (MAD) first introduced in 2003 and Low Glycemic Index Treatment (LGIT) first introduced in 2005 are another two dietary therapies, which are less restrictive, more palatable with fewer adverse effects and comparable efficacy. MAD is also a high fat, low carbohydrate diet, in which high sugar foods are discouraged and protein and fluids are unrestricted. In LGIT, only carbohydrates with Glycemic Index <50 are allowed and carbohydrate intake is restricted to 40-60 g per day. Medium Chain Triglyceride KD (MCT KD) is another alternative, in which there are more food choices as compared to classic KD, with comparable efficacy.


Assuntos
Dieta Cetogênica , Epilepsia Resistente a Medicamentos/dietoterapia , Criança , Dieta Cetogênica/efeitos adversos , Dieta Cetogênica/história , Gorduras na Dieta/administração & dosagem , Epilepsia Resistente a Medicamentos/metabolismo , Índice Glicêmico , História do Século XX , História do Século XXI , História Antiga , Humanos , Corpos Cetônicos/biossíntese , Triglicerídeos/administração & dosagem
17.
EMBO Mol Med ; 10(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29976786

RESUMO

Sepsis is a highly lethal and urgent unmet medical need. It is the result of a complex interplay of several pathways, including inflammation, immune activation, hypoxia, and metabolic reprogramming. Specifically, the regulation and the impact of the latter have become better understood in which the highly catabolic status during sepsis and its similarity with starvation responses appear to be essential in the poor prognosis in sepsis. It seems logical that new interventions based on the recognition of new therapeutic targets in the key metabolic pathways should be developed and may have a good chance to penetrate to the bedside. In this review, we concentrate on the pathological changes in metabolism, observed during sepsis, and the presumed underlying mechanisms, with a focus on the level of the organism and the interplay between different organ systems.


Assuntos
Sepse/metabolismo , Animais , Glicemia/metabolismo , Epigênese Genética , Glicólise , Humanos , Hiperglicemia/metabolismo , Hipoglicemia/metabolismo , Hipóxia/metabolismo , Inflamação/metabolismo , Corpos Cetônicos/biossíntese , Lipólise , Redes e Vias Metabólicas , Camundongos , Mitocôndrias/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Ratos , Sepse/sangue , Sepse/diagnóstico , Sepse/genética
18.
Endocrinology ; 159(5): 2110-2126, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29596655

RESUMO

The adaptive fasting response is invoked as a promising cardiometabolic and neurodegenerative therapeutic pathway. We and others have defined the carbohydrate transporter glucose transporter 8 (GLUT8) as a critical regulator of hepatic and whole-organism metabolic homeostasis in the overfed and diabetic states. However, the functions of this critical transporter in the physiological fasting response remain poorly understood. Here, we tested the hypothesis that GLUT8 modulates the adaptive hepatic fasting response. We demonstrate that mice with targeted Slc2a8 disruption exhibit enhanced thermogenesis, ketogenesis, and peripheral lipid mobilization during fasting. These metabolic enhancements were observed in the context of mildly impaired hepatic mitochondrial oxidative metabolism in vivo and in vitro. Mechanistically, we show that hepatic peroxisome proliferator-activated receptor α (PPARα) and its transcriptional fasting response target hepatokine, fibroblast growth factor (FGF)21, are cell-autonomously hyperactivated in GLUT8-deficient liver and in isolated primary murine hepatocytes during nutrient depletion. Hepatic PPARα knockdown in GLUT8-deficient mice normalized the enhanced ketogenic and FGF21 secretory responses and decreased mitochondrial respiratory function without altering the hyperthermic response to fasting. Our data demonstrate that hepatocyte GLUT8 regulates adaptive fasting in part through regulation of the PPARα signaling cascade. Moreover, the ketotic and thermic responses to fasting are differentially encoded within the GLUT8-PPARα communication axis. GLUT8 therefore represents a therapeutic target that can be leveraged against cardiometabolic disease.


Assuntos
Jejum/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Proteínas Facilitadoras de Transporte de Glucose/genética , Hepatócitos/metabolismo , Corpos Cetônicos/biossíntese , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , PPAR alfa/metabolismo , Animais , Técnicas de Silenciamento de Genes , Proteínas Facilitadoras de Transporte de Glucose/deficiência , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Transdução de Sinais , Termogênese/genética
19.
Sci Rep ; 7(1): 4590, 2017 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-28676675

RESUMO

Diabetes increases the risk of Cardio-vascular disease (CVD). CVD is more prevalent in type 2 diabetes (T2D) than type 1 diabetes (T1D), but the mortality risk is higher in T1D than in T2D. The pathophysiology of CVD in T1D is poorly defined. To learn more about biological pathways that are potentially involved in T1D with cardiac dysfunction, we sought to identify differentially expressed genes in the T1D heart. Our study used T1D mice with severe hyperglycemia along with significant deficits in echocardiographic measurements. Microarray analysis of heart tissue RNA revealed that the T1D mice differentially expressed 10 genes compared to control. Using Ingenuity Pathway Analysis (IPA), we showed that these genes were significantly involved in ketogenesis, cardiovascular disease, apoptosis and other toxicology functions. Of these 10 genes, the 3-Hydroxy-3-Methylglutaryl-CoA Synthase 2 (HMGCS2) was the highest upregulated gene in T1D heart. IPA analysis showed that HMGCS2 was center to many biological networks and pathways. Our data also suggested that apart from heart, the expression of HMGCS2 was also different in kidney and spleen between control and STZ treated mice. In conclusion, The HMGCS2 molecule may potentially be involved in T1D induced cardiac dysfunction.


Assuntos
Doenças Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/biossíntese , Animais , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/fisiopatologia , Biologia Computacional/métodos , Diabetes Mellitus Tipo 1/genética , Ecocardiografia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Hemodinâmica , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Masculino , Camundongos , Anotação de Sequência Molecular , Transcriptoma
20.
J Am Heart Assoc ; 6(4)2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28381467

RESUMO

BACKGROUND: Ketone bodies are known to substitute for glucose as brain fuel when glucose availability is low. Ketogenic diets have been described as neuroprotective. Similar data have been reported for triheptanoin, a fatty oil and anaplerotic compound. In this study, we monitored the changes of energy metabolites in liver, blood, and brain after transient brain ischemia to test for ketone body formation induced by experimental stroke. METHODS AND RESULTS: Mice were fed a standard carbohydrate-rich diet or 2 fat-rich diets, 1 enriched in triheptanoin and 1 in soybean oil. Stroke was induced in mice by middle cerebral artery occlusion for 90 minutes, followed by reperfusion. Mice were sacrificed, and blood plasma and liver and brain homogenates were obtained. In 1 experiment, microdialysis was performed. Metabolites (eg glucose, ß-hydroxybutyrate, citrate, succinate) were determined by gas chromatography-mass spectrometry. After 90 minutes of brain ischemia, ß-hydroxybutyrate levels were dramatically increased in liver, blood, and brain microdialysate and brain homogenate, but only in mice fed fat-rich diets. Glucose levels were changed in the opposite manner in blood and brain. Reperfusion decreased ß-hydroxybutyrate and increased glucose within 60 minutes. Stroke-induced ketogenesis was blocked by propranolol, a ß-receptor antagonist. Citrate and succinate were moderately increased by fat-rich diets and unchanged after stroke. CONCLUSIONS: We conclude that brain ischemia induces the formation of ß-hydroxybutyrate (ketogenesis) in the liver and the consumption of ß-hydroxybutyrate in the brain. This effect seems to be mediated by ß-adrenergic receptors.


Assuntos
Encéfalo/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Corpos Cetônicos/biossíntese , Fígado/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Isquemia Encefálica/metabolismo , Ácido Cítrico/metabolismo , Dieta da Carga de Carboidratos , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Glucose/metabolismo , Fígado/efeitos dos fármacos , Camundongos , Microdiálise , Propranolol/farmacologia , Óleo de Soja , Ácido Succínico/metabolismo , Triglicerídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...