Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 36(5): 1088-1104.e12, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38447582

RESUMEN

Acetyl-CoA carboxylase (ACC) promotes prandial liver metabolism by producing malonyl-CoA, a substrate for de novo lipogenesis and an inhibitor of CPT-1-mediated fat oxidation. We report that inhibition of ACC also produces unexpected secondary effects on metabolism. Liver-specific double ACC1/2 knockout (LDKO) or pharmacologic inhibition of ACC increased anaplerosis, tricarboxylic acid (TCA) cycle intermediates, and gluconeogenesis by activating hepatic CPT-1 and pyruvate carboxylase flux in the fed state. Fasting should have marginalized the role of ACC, but LDKO mice maintained elevated TCA cycle intermediates and preserved glycemia during fasting. These effects were accompanied by a compensatory induction of proteolysis and increased amino acid supply for gluconeogenesis, which was offset by increased protein synthesis during feeding. Such adaptations may be related to Nrf2 activity, which was induced by ACC inhibition and correlated with fasting amino acids. The findings reveal unexpected roles for malonyl-CoA synthesis in liver and provide insight into the broader effects of pharmacologic ACC inhibition.


Asunto(s)
Acetil-CoA Carboxilasa , Aminoácidos , Gluconeogénesis , Hígado , Malonil Coenzima A , Ratones Noqueados , Oxidación-Reducción , Animales , Malonil Coenzima A/metabolismo , Hígado/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Ratones , Aminoácidos/metabolismo , Masculino , Piruvato Carboxilasa/metabolismo , Ciclo del Ácido Cítrico , Ácido Pirúvico/metabolismo , Ratones Endogámicos C57BL , Ayuno/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo
2.
J Clin Invest ; 133(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36928190

RESUMEN

BACKGROUNDHepatic de novo lipogenesis (DNL) and ß-oxidation are tightly coordinated, and their dysregulation is thought to contribute to the pathogenesis of nonalcoholic fatty liver (NAFL). Fasting normally relaxes DNL-mediated inhibition of hepatic ß-oxidation, dramatically increasing ketogenesis and decreasing reliance on the TCA cycle. Thus, we tested whether aberrant oxidative metabolism in fasting NAFL subjects is related to the inability to halt fasting DNL.METHODSForty consecutive nondiabetic individuals with and without a history of NAFL were recruited for this observational study. After phenotyping, subjects fasted for 24 hours, and hepatic metabolism was interrogated using a combination of 2H2O and 13C tracers, magnetic resonance spectroscopy, and high-resolution mass spectrometry.RESULTSWithin a subset of subjects, DNL was detectable after a 24-hour fast and was more prominent in those with NAFL, though it was poorly correlated with steatosis. However, fasting DNL negatively correlated with hepatic ß-oxidation and ketogenesis and positively correlated with citrate synthesis. Subjects with NAFL but undetectable fasting DNL (25th percentile) were comparatively normal. However, those with the highest fasting DNL (75th percentile) were intransigent to the effects of fasting on the concentration of insulin, non-esterified fatty acid, and ketones. Additionally, they sustained glycogenolysis and were spared the loss of oxaloacetate to gluconeogenesis in favor of citrate synthesis, which correlated with DNL and diminished ketogenesis.CONCLUSIONMetabolic flux analysis in fasted subjects indicates that shared metabolic mechanisms link the dysregulations of hepatic DNL, ketogenesis, and the TCA cycle in NAFL.TRIAL REGISTRATIONData were obtained during the enrollment/non-intervention phase of Effect of Vitamin E on Non-Alcoholic Fatty Liver Disease, ClinicalTrials.gov NCT02690792.FUNDINGThis work was supported by the University of Texas Southwestern NORC Quantitative Metabolism Core (NIH P30DK127984), the NIH/National Institute of Diabetes and Digestive and Kidney Diseases (R01DK078184, R01DK128168, R01DK087977, R01DK132254, and K01DK133630), the NIH/National Institute on Alcohol Abuse and Alcoholism (K01AA030327), and the Robert A. Welch Foundation (I-1804).


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Lipogénesis/fisiología , Ácido Cítrico , Hígado/metabolismo , Cuerpos Cetónicos/metabolismo , Citratos/metabolismo , Ayuno
4.
Cell Rep ; 39(4): 110733, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476997

RESUMEN

Hepatic gluconeogenesis from amino acids contributes significantly to diabetic hyperglycemia, but the molecular mechanisms involved are incompletely understood. Alanine transaminases (ALT1 and ALT2) catalyze the interconversion of alanine and pyruvate, which is required for gluconeogenesis from alanine. We find that ALT2 is overexpressed in the liver of diet-induced obese and db/db mice and that the expression of the gene encoding ALT2 (GPT2) is downregulated following bariatric surgery in people with obesity. The increased hepatic expression of Gpt2 in db/db liver is mediated by activating transcription factor 4, an endoplasmic reticulum stress-activated transcription factor. Hepatocyte-specific knockout of Gpt2 attenuates incorporation of 13C-alanine into newly synthesized glucose by hepatocytes. In vivo Gpt2 knockdown or knockout in liver has no effect on glucose concentrations in lean mice, but Gpt2 suppression alleviates hyperglycemia in db/db mice. These data suggest that ALT2 plays a significant role in hepatic gluconeogenesis from amino acids in diabetes.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Alanina/farmacología , Alanina Transaminasa/metabolismo , Aminoácidos/metabolismo , Animales , Diabetes Mellitus/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Obesidad/metabolismo
5.
Cell Mol Gastroenterol Hepatol ; 13(3): 879-899, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34923175

RESUMEN

BACKGROUND & AIMS: Substitution of lysine for glutamic acid at residu 167 in Transmembrane 6 superfamily member 2 (TM6SF2) is associated with fatty liver disease and reduced plasma lipid levels. Tm6sf2-/- mice replicate the human phenotype but were not suitable for detailed mechanistic studies. As an alternative model, we generated Tm6sf2-/- rats to determine the subcellular location and function of TM6SF2. METHODS: Two lines of Tm6sf2-/- rats were established using gene editing. Lipids from tissues and from newly secreted very low density lipoproteins (VLDLs) were quantified using enzymatic assays and mass spectrometry. Neutral lipids were visualized in tissue sections using Oil Red O staining. The rate of dietary triglyceride (TG) absorption and hepatic VLDL-TG secretion were compared in Tm6sf2-/- mice and in their wild-type littermates. The intracellular location of TM6SF2 was determined by cell fractionation. Finally, TM6SF2 was immunoprecipitated from liver and enterocytes to identify interacting proteins. RESULTS: Tm6sf2-/- rats had a 6-fold higher mean hepatic TG content (56.1 ± 28.9 9 vs 9.8 ± 3.9 mg/g; P < .0001) and lower plasma cholesterol levels (99.0 ± 10.5 vs 110.6 ± 14.0 mg/dL; P = .0294) than their wild-type littermates. Rates of appearance of dietary and hepatic TG into blood were reduced significantly in Tm6sf2-/- rats (P < .001 and P < .01, respectively). Lipid content of newly secreted VLDLs isolated from perfused livers was reduced by 53% (TG) and 62% (cholesterol) (P = .005 and P = .01, respectively) in Tm6sf2-/- mice. TM6SF2 was present predominantly in the smooth endoplasmic reticulum and endoplasmic reticulum-Golgi intermediate compartments, but not in Golgi. Both apolipoprotein B-48 and acyl-CoA synthetase long chain family member 5 physically interacted with TM6SF2. CONCLUSIONS: TM6SF2 acts in the smooth endoplasmic reticulum to promote bulk lipidation of apolipoprotein B-containing lipoproteins, thus preventing fatty liver disease.


Asunto(s)
Proteínas de la Membrana , Enfermedad del Hígado Graso no Alcohólico , Animales , Proteínas de la Membrana/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratas
6.
Nat Commun ; 12(1): 3756, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34145255

RESUMEN

De novo lipogenesis (DNL) is disrupted in a wide range of human disease. Thus, quantification of DNL may provide insight into mechanisms and guide interventions if it can be performed rapidly and noninvasively. DNL flux is commonly measured by 2H incorporation into fatty acids following deuterated water (2H2O) administration. However, the sensitivity of this approach is limited by the natural abundance of 13C, which masks detection of 2H by mass spectrometry. Here we report that high-resolution Orbitrap gas-chromatography mass-spectrometry resolves 2H and 13C fatty acid mass isotopomers, allowing DNL to be quantified using lower 2H2O doses and shorter experimental periods than previously possible. Serial measurements over 24-hrs in mice detects the nocturnal activation of DNL and matches a 3H-water method in mice with genetic activation of DNL. Most importantly, DNL is detected in overnight-fasted humans in less than an hour and is responsive to feeding during a 4-h study. Thus, 2H specific MS provides the ability to study DNL in settings that are currently impractical.


Asunto(s)
Ácidos Grasos/biosíntesis , Cromatografía de Gases y Espectrometría de Masas/métodos , Lipogénesis/fisiología , Hígado/metabolismo , Triglicéridos/biosíntesis , Animales , Deuterio/química , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Metab Eng ; 59: 1-14, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31891762

RESUMEN

Computational models based on the metabolism of stable isotope tracers can yield valuable insight into the metabolic basis of disease. The complexity of these models is limited by the number of tracers and the ability to characterize tracer labeling in downstream metabolites. NMR spectroscopy is ideal for multiple tracer experiments since it precisely detects the position of tracer nuclei in molecules, but it lacks sensitivity for detecting low-concentration metabolites. GC-MS detects stable isotope mass enrichment in low-concentration metabolites, but lacks nuclei and positional specificity. We performed liver perfusions and in vivo infusions of 2H and 13C tracers, yielding complex glucose isotopomers that were assigned by NMR and fit to a newly developed metabolic model. Fluxes regressed from 2H and 13C NMR positional isotopomer enrichments served to validate GC-MS-based flux estimates obtained from the same experimental samples. NMR-derived fluxes were largely recapitulated by modeling the mass isotopomer distributions of six glucose fragment ions measured by GC-MS. Modest differences related to limited fragmentation coverage of glucose C1-C3 were identified, but fluxes such as gluconeogenesis, glycogenolysis, cataplerosis and TCA cycle flux were tightly correlated between the methods. Most importantly, modeling of GC-MS data could assign fluxes in primary mouse hepatocytes, an experiment that is impractical by 2H or 13C NMR.


Asunto(s)
Ciclo del Ácido Cítrico , Gluconeogénesis , Hígado/metabolismo , Modelos Biológicos , Vía de Pentosa Fosfato , Animales , Isótopos de Carbono/análisis , Isótopos de Carbono/química , Isótopos de Carbono/farmacología , Masculino , Ratones , Resonancia Magnética Nuclear Biomolecular
8.
JCI Insight ; 52019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31012869

RESUMEN

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.


Asunto(s)
Acetilcoenzima A/metabolismo , Hiperglucemia/metabolismo , Cuerpos Cetónicos/biosíntesis , Cetosis/complicaciones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Adulto , Glucemia/análisis , Ciclo del Ácido Cítrico , Metabolismo Energético , Ayuno/fisiología , Femenino , Gluconeogénesis , Técnica de Clampeo de la Glucosa , Humanos , Hiperglucemia/sangre , Hiperglucemia/etiología , Cuerpos Cetónicos/análisis , Cetosis/sangre , Cetosis/metabolismo , Cetosis/fisiopatología , Hígado/diagnóstico por imagen , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Espectroscopía de Protones por Resonancia Magnética , Triglicéridos/análisis , Triglicéridos/metabolismo
9.
Cell Metab ; 29(6): 1291-1305.e8, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31006591

RESUMEN

The hepatic TCA cycle supports oxidative and biosynthetic metabolism. This dual responsibility requires anaplerotic pathways, such as pyruvate carboxylase (PC), to generate TCA cycle intermediates necessary for biosynthesis without disrupting oxidative metabolism. Liver-specific PC knockout (LPCKO) mice were created to test the role of anaplerotic flux in liver metabolism. LPCKO mice have impaired hepatic anaplerosis, diminution of TCA cycle intermediates, suppressed gluconeogenesis, reduced TCA cycle flux, and a compensatory increase in ketogenesis and renal gluconeogenesis. Loss of PC depleted aspartate and compromised urea cycle function, causing elevated urea cycle intermediates and hyperammonemia. Loss of PC prevented diet-induced hyperglycemia and insulin resistance but depleted NADPH and glutathione, which exacerbated oxidative stress and correlated with elevated liver inflammation. Thus, despite catalyzing the synthesis of intermediates also produced by other anaplerotic pathways, PC is specifically necessary for maintaining oxidation, biosynthesis, and pathways distal to the TCA cycle, such as antioxidant defenses.


Asunto(s)
Antioxidantes/metabolismo , Ciclo del Ácido Cítrico/genética , Hígado/metabolismo , Redes y Vías Metabólicas/genética , Piruvato Carboxilasa/genética , Animales , Respiración de la Célula/genética , Gluconeogénesis/genética , Hepatitis/genética , Hepatitis/metabolismo , Hepatitis/patología , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/metabolismo , Oxidación-Reducción , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Piruvato Carboxilasa/metabolismo
10.
Exp Physiol ; 103(3): 408-418, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29215172

RESUMEN

NEW FINDINGS: What is the central question of this study? Does a reduction in hepatic peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), which has been observed in an insulin-resistant obese state, impair the ability of fibroblast growth factor 21 (FGF21) to modulate metabolism? What is the main finding and its importance? A deficit in hepatic PGC-1α does not compromise the ability of FGF21 to increase hepatic fatty acid oxidation; however, the effects of FGF21 to regulate whole-body metabolism (i.e. total and resting energy expenditure), as well as ambulatory activity, were altered when hepatic PGC-1α was reduced. ABSTRACT: Fibroblast growth factor 21 (FGF21) treatment drives metabolic improvements, including increased metabolic flux and reduced hepatic steatosis, but the mechanisms responsible for these effects remain to be elucidated fully. We tested whether a targeted reduction in hepatic peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), which has been shown to occur with obesity, had a negative impact on the metabolic effects of FGF21. We infused FGF21 (1 mg kg-1  day-1 ) or saline in chow-fed wild-type (WT) and liver-specific PGC-1α heterozygous (LPGC-1α) mice for 4 weeks. Administration of FGF21 lowered serum insulin and cholesterol (P ≤ 0.05) and tended to lower free fatty acids (P = 0.057). The LPGC-1α mice exhibited reduced complete hepatic fatty acid oxidation (FAO; LPGC-1α, 1788 ± 165 nmol g-1  h-1 compared with WT, 2572 ± 437 nmol g-1  h-1 ; P < 0.001), which was normalized by FGF21 treatment (2788 ± 519 nmol g-1  h-1 ; P < 0.001). FGF21 also increased hepatic incomplete FAO by 12% in both groups and extramitochondrial FAO by 89 and 56% in WT and LPGC-1α mice, respectfully (P = 0.001), and lowered hepatic triacylglycerol by 30-40% (P < 0.001). Chronic treatment with FGF21 lowered body weight and fat mass (P < 0.05), while increasing food consumption (P < 0.05), total energy expenditure [7.3 ± 0.60 versus 6.6 ± 0.39 kcal (12 h)-1 in WT mice; P = 0.009] and resting energy expenditure [5.4 ± 0.89 versus 4.6 ± 0.21 kcal (12 h)-1 in WT mice; P = 0.005]. Interestingly, FGF21 only increased ambulatory activity in the WT mice (P = 0.03), without a concomitant increase in non-resting energy expenditure. In conclusion, although reduced hepatic PGC-1α expression was not necessary for FGF21 to increase FAO, it does appear to mediate FGF21-induced changes in total and resting energy expenditure and ambulatory activity in lean mice.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Hígado/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Colesterol/sangre , Ácidos Grasos no Esterificados/sangre , Insulina/sangre , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Noqueados , Oxidación-Reducción , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética
11.
J Endocrinol ; 235(2): 97-109, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28765264

RESUMEN

Exercise enhances insulin sensitivity; it also improves adipocyte metabolism and reduces adipose tissue inflammation through poorly defined mechanisms. Fibroblast growth factor 21 (FGF21) is a pleiotropic hormone-like protein whose insulin-sensitizing properties are predominantly mediated via receptor signaling in adipose tissue (AT). Recently, FGF21 has also been demonstrated to have anti-inflammatory properties. Meanwhile, an association between exercise and increased circulating FGF21 levels has been reported in some, but not all studies. Thus, the role that FGF21 plays in mediating the positive metabolic effects of exercise in AT are unclear. In this study, FGF21-knockout (KO) mice were used to directly assess the role of FGF21 in mediating the metabolic and anti-inflammatory effects of exercise on white AT (WAT) and brown AT (BAT). Male FGF21KO and wild-type mice were provided running wheels or remained sedentary for 8 weeks (n = 9-15/group) and compared for adiposity, insulin sensitivity (i.e., HOMA-IR, Adipo-IR) and AT inflammation and metabolic function (e.g., mitochondrial enzyme activity, subunit content). Adiposity and Adipo-IR were increased in FGF21KO mice and decreased by EX. The BAT of FGF21KO animals had reduced mitochondrial content and decreased relative mass, both normalized by EX. WAT and BAT inflammation was elevated in FGF21KO mice, reduced in both genotypes by EX. EX increased WAT Pgc1alpha gene expression, citrate synthase activity, COX I content and total AMPK content in WT but not FGF21KO mice. Collectively, these findings reveal a previously unappreciated anti-inflammatory role for FGF21 in WAT and BAT, but do not support that FGF21 is necessary for EX-mediated anti-inflammatory effects.


Asunto(s)
Tejido Adiposo/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Inflamación/metabolismo , Condicionamiento Físico Animal/fisiología , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/fisiología , Genotipo , Masculino , Ratones , Ratones Noqueados
12.
J Physiol ; 595(14): 4909-4926, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28504310

RESUMEN

KEY POINTS: Low intrinsic aerobic capacity is associated with increased all-cause and liver-related mortality in humans. Low intrinsic aerobic capacity in the low capacity runner (LCR) rat increases susceptibility to acute and chronic high-fat/high-sucrose diet-induced steatosis, without observed increases in liver inflammation. Addition of excess cholesterol to a high-fat/high-sucrose diet produced greater steatosis in LCR and high capacity runner (HCR) rats. However, the LCR rat demonstrated greater susceptibility to increased liver inflammatory and apoptotic markers compared to the HCR rat. The progressive non-alcoholic fatty liver disease observed in the LCR rats following western diet feeding was associated with further declines in liver fatty acid oxidation and mitochondrial respiratory capacity compared to HCR rats. ABSTRACT: Low aerobic capacity increases risk for non-alcoholic fatty liver disease and liver-related disease mortality, but mechanisms mediating these effects remain unknown. We recently reported that rats bred for low aerobic capacity (low capacity runner; LCR) displayed susceptibility to high fat diet-induced steatosis in association with reduced hepatic mitochondrial fatty acid oxidation (FAO) and respiratory capacity compared to high aerobic capacity (high capacity runner; HCR) rats. Here we tested the impact of aerobic capacity on susceptibility for progressive liver disease following a 16-week 'western diet' (WD) high in fat (45% kcal), cholesterol (1% w/w) and sucrose (15% kcal). Unlike previously with a diet high in fat and sucrose alone, the inclusion of cholesterol in the WD induced hepatomegaly and steatosis in both HCR and LCR rats, while producing greater cholesterol ester accumulation in LCR compared to HCR rats. Importantly, WD-fed low-fitness LCR rats displayed greater inflammatory cell infiltration, serum alanine transaminase, expression of hepatic inflammatory markers (F4/80, MCP-1, TLR4, TLR2 and IL-1ß) and effector caspase (caspase 3 and 7) activation compared to HCR rats. Further, LCR rats had greater WD-induced decreases in complete FAO and mitochondrial respiratory capacity. Intrinsic aerobic capacity had no impact on WD-induced hepatic steatosis; however, rats bred for low aerobic capacity developed greater hepatic inflammation, which was associated with reduced hepatic mitochondrial FAO and respiratory capacity and increased accumulation of cholesterol esters. These results confirm epidemiological reports that aerobic capacity impacts progression of liver disease and suggest that these effects are mediated through alterations in hepatic mitochondrial function.


Asunto(s)
Dieta , Hígado Graso/metabolismo , Hígado Graso/patología , Carrera/fisiología , Animales , Colesterol/metabolismo , Citrato (si)-Sintasa/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Masculino , Mitocondrias Hepáticas/metabolismo , Oxidación-Reducción , Palmitatos/metabolismo , Ácido Pirúvico/metabolismo , Ratas
13.
Am J Physiol Endocrinol Metab ; 311(4): E749-E760, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27600823

RESUMEN

Rats selectively bred for high capacity running (HCR) or low capacity running (LCR) display divergence for intrinsic aerobic capacity and hepatic mitochondrial oxidative capacity, both factors associated with susceptibility for nonalcoholic fatty liver disease. Here, we tested if HCR and LCR rats display differences in susceptibility for hepatic steatosis after 16 wk of high-fat diets (HFD) with either 45% or 60% of kcals from fat. HCR rats were protected against HFD-induced hepatic steatosis, whereas only the 60% HFD induced steatosis in LCR rats, as marked by a doubling of liver triglycerides. Hepatic complete fatty acid oxidation (FAO) and mitochondrial respiratory capacity were all lower in LCR compared with HCR rats. LCR rats also displayed lower hepatic complete and incomplete FAO in the presence of etomoxir, suggesting a reduced role for noncarnitine palmitoyltransferase-1-mediated lipid catabolism in LCR versus HCR rats. Hepatic complete FAO and mitochondrial respiration were largely unaffected by either chronic HFD; however, 60% HFD feeding markedly reduced 2-pyruvate oxidation, a marker of tricarboxylic acid (TCA) cycle flux, and mitochondrial complete FAO only in LCR rats. LCR rats displayed lower levels of hepatic long-chain acylcarnitines than HCR rats but maintained similar levels of hepatic acetyl-carnitine levels, further supporting lower rates of ß-oxidation, and TCA cycle flux in LCR than HCR rats. Finally, only LCR rats displayed early reductions in TCA cycle genes after the acute initiation of a HFD. In conclusion, intrinsically high aerobic capacity confers protection against HFD-induced hepatic steatosis through elevated hepatic mitochondrial oxidative capacity.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Mitocondrias Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Aerobiosis , Animales , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/metabolismo , Ciclo del Ácido Cítrico/efectos de los fármacos , Dieta Alta en Grasa , Susceptibilidad a Enfermedades , Ácidos Grasos/metabolismo , Hígado/metabolismo , Masculino , Oxidación-Reducción , Estrés Oxidativo , Ácido Pirúvico/metabolismo , Ratas , Triglicéridos/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G387-95, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27445343

RESUMEN

Hyperphagic Otsuka Long-Evans Tokushima fatty (OLETF) rats develop obesity, insulin resistance, and nonalcoholic fatty liver disease (NAFLD), but lifestyle modifications, such as caloric restriction (CR), can prevent these conditions. We sought to determine if prior CR had protective effects on metabolic health and NAFLD development following a 4-wk return to ad libitum (AL) feeding. Four-week-old male OLETF rats (n = 8-10/group) were fed AL for 16 wk (O-AL), CR for 16 wk (O-CR; ∼70% kcal of O-AL), or CR for 12 wk followed by 4 wk of AL feeding (O-AL4wk). CR-induced benefit in prevention of NAFLD, including reduced hepatic steatosis, inflammation, and markers of Kupffer cell activation/number, was largely lost in AL4wk rats. These findings occurred in conjunction with a partial loss of CR-induced beneficial effects on obesity and serum triglycerides in O-AL4wk rats, but in the absence of changes in serum glucose or insulin. CR-induced increases in hepatic mitochondrial respiration remained significantly elevated (P < 0.01) in O-AL4wk compared with O-AL rats, while mitochondrial [1-(14)C]palmitate oxidation, citrate synthase activity, and ß-hydroxyacyl-CoA dehydrogenase activity did not differ among OLETF groups. NAFLD development in O-AL4wk rats was accompanied by increases in the protein content of the de novo lipogenesis markers fatty acid synthase and stearoyl-CoA desaturase-1 and decreases in phosphorylated acetyl-CoA carboxylase (pACC)/ACC compared with O-CR rats (P < 0.05 for each). The beneficial effects of chronic CR on NAFLD development were largely lost with 4 wk of AL feeding in the hyperphagic OLETF rat, highlighting the importance of maintaining energy balance in the prevention of NAFLD.


Asunto(s)
Restricción Calórica , Hígado Graso/dietoterapia , Animales , Biomarcadores , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos , Lipogénesis , Masculino , Mitocondrias Hepáticas/metabolismo , Ratas , Ratas Endogámicas OLETF
16.
Am J Physiol Gastrointest Liver Physiol ; 310(10): G832-43, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27012775

RESUMEN

Exercise stimulates hepatic mitochondrial adaptations; however, the mechanisms remain largely unknown. Here we tested whether FGF21 plays an obligatory role in exercise induced hepatic mitochondrial adaptations by testing exercise responses in FGF21 knockout mice. FGF21 knockout (FGF21-KO) and wild-type (WT) mice (11-12 wk of age) had access to voluntary running wheels for exercise (EX) or remained sedentary for 8 wk. FGF21 deficiency resulted in greater body weight, adiposity, serum cholesterol, insulin, and glucose concentrations compared with WT mice (P < 0.05). In addition, hepatic mitochondrial complete palmitate oxidation, ß-hydroxyacyl-CoA dehydrogenase (ß-HAD) activity, and nuclear content of PGC-1α were 30-50% lower in FGF21-KO mice compared with WT mice (P < 0.01). EX effectively lowered body weight, adiposity, serum triglycerides, free fatty acids, and insulin and normalized mitochondrial complete palmitate oxidation in the FGF21-KO mice, whereas the reduced hepatic ß-HAD activity and lowered nuclear content of PGC-1α in FGF21-KO mice were not restored by EX. In addition, EX increased hepatic CPT-1α mRNA expression and ACC phosphorylation (a marker of increased AMPK activity) and reduced hepatic triacylglycerol content in both genotypes. However, FGF21-KO mice displayed a lower EX-induced increase in the mRNA expression of the hepatic gluconeogenic gene, PEPCK, compared with WT. In conclusion, FGF21 does not appear necessary for exercise-induced systemic and hepatic mitochondrial adaptations, but the increased adiposity, hyperinsulinemia, and impairments in hepatic mitochondrial function induced by FGF21 deficiency can be partially rescued by daily wheel running exercise.


Asunto(s)
Adaptación Fisiológica , Factores de Crecimiento de Fibroblastos/genética , Mitocondrias Hepáticas/metabolismo , Carrera , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Composición Corporal , Carnitina O-Palmitoiltransferasa/metabolismo , Colesterol/sangre , Factores de Crecimiento de Fibroblastos/metabolismo , Gluconeogénesis , Insulina/sangre , Ratones , Ratones Endogámicos C57BL , Palmitatos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo
17.
J Hepatol ; 64(1): 171-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26325536

RESUMEN

BACKGROUND & AIMS: Mounting evidence indicates that maternal exercise confers protection to adult offspring against various diseases. Here we hypothesized that maternal exercise during gestation would reduce high-fat diet (HFD)-induced hepatic steatosis in adult rat offspring. METHODS: Following conception, pregnant dams were divided into either voluntary wheel running exercise (GE) or wheel-locked sedentary (GS) groups throughout gestation (days 4-21). Post-weaning, offspring received either normal chow diet (CD; 10% fat, 70% carbohydrate, 20% protein) or HFD (45% fat, 35% carbohydrate, and 20% protein) until sacrificed at 4- or 8-months of age. RESULTS: GE did not affect offspring birth weight or litter size. HFD feeding in offspring increased weight gain, body fat percentage, and glucose tolerance test area under the curve (GTT-AUC). Male offspring from GE dams had reduced body fat percentage across all ages (p<0.05). In addition, 8-month male offspring from GE dams were protected against HFD-induced hepatic steatosis, which was associated with increased markers of hepatic mitochondrial biogenesis (PGC-1α and TFAM), autophagic potential (ATG12:ATG5 conjugation) and hepatic triacylglycerol secretion (MTTP). CONCLUSIONS: The current study provides the first evidence that gestational exercise can reduce susceptibility to HFD-induced hepatic steatosis in adult male offspring.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/prevención & control , Condicionamiento Físico Animal , Animales , Proteínas Portadoras/análisis , Dieta Alta en Grasa , Femenino , Masculino , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Embarazo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/análisis
18.
J Clin Invest ; 125(12): 4447-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26571396

RESUMEN

Mitochondria are critical for respiration in all tissues; however, in liver, these organelles also accommodate high-capacity anaplerotic/cataplerotic pathways that are essential to gluconeogenesis and other biosynthetic activities. During nonalcoholic fatty liver disease (NAFLD), mitochondria also produce ROS that damage hepatocytes, trigger inflammation, and contribute to insulin resistance. Here, we provide several lines of evidence indicating that induction of biosynthesis through hepatic anaplerotic/cataplerotic pathways is energetically backed by elevated oxidative metabolism and hence contributes to oxidative stress and inflammation during NAFLD. First, in murine livers, elevation of fatty acid delivery not only induced oxidative metabolism, but also amplified anaplerosis/cataplerosis and caused a proportional rise in oxidative stress and inflammation. Second, loss of anaplerosis/cataplerosis via genetic knockdown of phosphoenolpyruvate carboxykinase 1 (Pck1) prevented fatty acid-induced rise in oxidative flux, oxidative stress, and inflammation. Flux appeared to be regulated by redox state, energy charge, and metabolite concentration, which may also amplify antioxidant pathways. Third, preventing elevated oxidative metabolism with metformin also normalized hepatic anaplerosis/cataplerosis and reduced markers of inflammation. Finally, independent histological grades in human NAFLD biopsies were proportional to oxidative flux. Thus, hepatic oxidative stress and inflammation are associated with elevated oxidative metabolism during an obesogenic diet, and this link may be provoked by increased work through anabolic pathways.


Asunto(s)
Hepatocitos/metabolismo , Mitocondrias Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo , Animales , Hepatocitos/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Mitocondrias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Ratas , Ratas Wistar
19.
Appl Physiol Nutr Metab ; 40(10): 1038-47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26394261

RESUMEN

Weight loss is recommended for patients with nonalcoholic fatty liver disease (NAFLD), while metformin may lower liver enzymes in type 2 diabetics. Yet, the efficacy of the combination of weight loss and metformin in the treatment of NAFLD is unclear. We assessed the effects of metformin, caloric restriction, and their combination on NAFLD in diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Male OLETF rats (age 20 weeks; n = 6-8 per group) were fed ad libitum (AL), given metformin (300 mg·kg(-1)·day(-1); Met), calorically restricted (70% of AL; CR), or calorically restricted and given metformin (CR+Met) for 12 weeks. Met lowered adiposity compared with AL but not to the same magnitude as CR or CR+Met (p < 0.05). Although only CR improved fasting insulin and glucose, the combination of CR+Met was needed to improve post-challenge glucose tolerance. All treatments lowered hepatic triglycerides, but further improvements were observed in the CR groups (p < 0.05, Met vs. CR or CR+Met) and a further reduction in serum alanine aminotransferases was observed in CR+Met rats. CR lowered markers of hepatic de novo lipogenesis (fatty acid synthase, acetyl-CoA carboxylase (ACC), and stearoyl-CoA desaturase-1 (SCD-1)) and increased hepatic mitochondrial activity (palmitate oxidation and ß-hydroxyacyl CoA dehydrogenase (ß-HAD) activity). Changes were enhanced in the CR+Met group for ACC, SCD-1, ß-HAD, and the mitophagy marker BNIP3. Met decreased total hepatic mTOR content and inhibited mTOR complex 1, which may have contributed to Met-induced reductions in de novo lipogenesis. These findings in the OLETF rat suggest that the combination of caloric restriction and metformin may provide a more optimal approach than either treatment alone in the management of type 2 diabetes and NAFLD.


Asunto(s)
Restricción Calórica , Diabetes Mellitus Tipo 2/terapia , Metformina/farmacología , Enfermedad del Hígado Graso no Alcohólico/terapia , Obesidad/terapia , Animales , Western Blotting , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Hipoglucemiantes/farmacología , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Ratas , Ratas Long-Evans
20.
Med Sci Sports Exerc ; 47(3): 556-67, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24983336

RESUMEN

BACKGROUND: There is increasing use of high-intensity interval-type exercise training in the management of many lifestyle-related diseases. PURPOSE: This study aimed to test the hypothesis that vigorous-intensity interval exercise is as effective as traditional moderate-intensity aerobic exercise training for nonalcoholic fatty liver disease (NAFLD) outcomes in obese, Otsuka Long-Evans Tokushima Fatty (OLETF) rats. METHODS: OLETF rats (age, 20 wk; n = 8-10 per group) were assigned to sedentary (O-SED), moderate-intensity exercise training (O-MOD EX; 20 m·min(-1), 15% incline, 60 min·d(-1), 5 d·wk(-1) of treadmill running), or vigorous-intensity interval exercise training (O-VIG EX; 40 m·min(-1), 15% incline, 6 × 2.5 min bouts per day, 5 d·wk(-1) of treadmill running) groups for 12 wk. RESULTS: Both MOD EX and VIG EX effectively lowered hepatic triglycerides, serum alanine aminotransferase (ALT), perivenular fibrosis, and hepatic collagen 1α1 messenger RNA (mRNA) expression (vs O-SED, P < 0.05). In addition, both interventions increased hepatic mitochondrial markers (citrate synthase activity and fatty acid oxidation) and suppressed markers of de novo lipogenesis (fatty acid synthase, acetyl coenzyme A carboxylase, Elovl fatty acid elongase 6, and steroyl CoA desaturase-1), whereas only MOD EX increased hepatic mitochondrial Beta-hydroxyacyl-CoA dehydrogenase (ß-HAD) activity and hepatic triglyceride export marker apoB100 and lowered fatty acid transporter CD36 compared with O-SED. Moreover, whereas total hepatic macrophage population markers (CD68 and F4/80 mRNA) did not differ among groups, MOD EX and VIG EX lowered M1 macrophage polarization markers (CD11c, interleukin-1ß, and tumor necrosis factor α mRNA) and MOD EX increased M2 macrophage marker, CD206 mRNA, compared with O-SED. CONCLUSIONS: The accumulation of 15 min·d(-1) of VIG EX for 12 wk had similar effectiveness as 60 min·d(-1) of MOD EX in the management of NAFLD in OLETF rats. These findings may have important health outcome implications as we work to design better exercise training programs for patients with NAFLD.


Asunto(s)
Terapia por Ejercicio/métodos , Enfermedad del Hígado Graso no Alcohólico/terapia , Condicionamiento Físico Animal , Animales , Glucemia/metabolismo , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Glucólisis , Corazón/anatomía & histología , Lípidos/sangre , Lipogénesis , Hígado/metabolismo , Masculino , Mitocondrias Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Tamaño de los Órganos , ARN Mensajero/metabolismo , Ratas Endogámicas OLETF , Triglicéridos/metabolismo , Pérdida de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA