Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(4): 106355, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-36994079

RESUMO

Sarcopenia is a common and devastating condition in patients with chronic kidney disease (CKD). Here, we provide evidence that the kidney-muscle crosstalk in sarcopenia is mediated by reduced insulin sensitivity and the activation of the muscle-specific isoform of AMP deaminase, AMPD1. By using a high protein-based CKD model of sarcopenia in mice and differentiated human myotubes, we show that urea reduces insulin-dependent glucose and phosphate uptake by the skeletal muscle, thus contributing to the hyperphosphatemia observed in CKD whereas depleting intramuscular phosphate needed to restore energy and inhibit AMPD1. Hyperactivated AMPD1, in turn, aggravates the low energy state in the muscle by removing free adenosine monophosphate (AMP) and producing proinflammatory factors and uric acid which contribute to the progression of kidney disease. Our data provide molecular and metabolic evidence supporting the use of strategies aimed to improve insulin sensitivity and to block AMPD1 to prevent sarcopenia in subjects with CKD.

2.
JCI Insight ; 6(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33320834

RESUMO

Subjects with obesity frequently have elevated serum vasopressin levels, noted by measuring the stable analog, copeptin. Vasopressin acts primarily to reabsorb water via urinary concentration. However, fat is also a source of metabolic water, raising the possibility that vasopressin might have a role in fat accumulation. Fructose has also been reported to stimulate vasopressin. Here, we tested the hypothesis that fructose-induced metabolic syndrome is mediated by vasopressin. Orally administered fructose, glucose, or high-fructose corn syrup increased vasopressin (copeptin) concentrations and was mediated by fructokinase, an enzyme specific for fructose metabolism. Suppressing vasopressin with hydration both prevented and ameliorated fructose-induced metabolic syndrome. The vasopressin effects were mediated by the vasopressin 1b receptor (V1bR), as V1bR-KO mice were completely protected, whereas V1a-KO mice paradoxically showed worse metabolic syndrome. The mechanism is likely mediated in part by de novo expression of V1bR in the liver that amplifies fructokinase expression in response to fructose. Thus, our studies document a role for vasopressin in water conservation via the accumulation of fat as a source of metabolic water. Clinically, they also suggest that increased water intake may be a beneficial way to both prevent or treat metabolic syndrome.


Assuntos
Frutose/metabolismo , Síndrome Metabólica/metabolismo , Receptores de Vasopressinas/metabolismo , Vasopressinas/metabolismo , Animais , Modelos Animais de Doenças , Ingestão de Líquidos/fisiologia , Frutoquinases/metabolismo , Frutose/administração & dosagem , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Síndrome Metabólica/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Vasopressinas/deficiência , Receptores de Vasopressinas/genética , Vasopressinas/antagonistas & inibidores , Vasopressinas/biossíntese
3.
Am J Physiol Renal Physiol ; 317(4): F941-F948, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31411075

RESUMO

Obesity and metabolic syndrome are well-known risk factors for chronic kidney disease (CKD); however, less is known about the mechanism(s) by which metabolic syndrome might accelerate kidney disease. We hypothesized that metabolic syndrome should accelerate the development of kidney disease and that it might be associated with alterations in energy metabolism. We studied the pound mouse (which develops early metabolic syndrome due to a leptin receptor deletion) and wild-type littermates and compared the level of renal injury and muscle wasting after equivalent injury with oral adenine. Renal function, histology, and biochemical analyses were performed. The presence of metabolic syndrome was associated with earlier development of renal disease (12 mo) and earlier mortality in pound mice compared with controls. After administration of adenine, kidney disease was worse in pound mice, and this was associated with greater tubular injury with a decrease in kidney mitochondria, lower tissue ATP levels, and worse oxidative stress. Pound mice with similar levels of renal function as adenine-treated wild-type mice also showed worse sarcopenia, with lower tissue ATP and intracellular phosphate levels. In summary, our data demonstrate that obesity and metabolic syndrome accelerate the progression of CKD and worsen CKD-dependent sarcopenia. Both conditions are associated with renal alterations in energy metabolism and lower tissue ATP levels secondary to mitochondrial dysfunction and reduced mitochondrial number.


Assuntos
Metabolismo Energético , Rim/metabolismo , Mitocôndrias/metabolismo , Obesidade/complicações , Obesidade/metabolismo , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Adenina/toxicidade , Trifosfato de Adenosina/metabolismo , Animais , Testes de Função Renal , Túbulos Renais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Sarcopenia/etiologia , Sarcopenia/metabolismo
4.
J Biol Chem ; 294(11): 4272-4281, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30651350

RESUMO

Dietary, fructose-containing sugars have been strongly associated with the development of nonalcoholic fatty liver disease (NAFLD). Recent studies suggest that fructose also can be produced via the polyol pathway in the liver, where it may induce hepatic fat accumulation. Moreover, fructose metabolism yields uric acid, which is highly associated with NAFLD. Here, using biochemical assays, reporter gene expression, and confocal fluorescence microscopy, we investigated whether uric acid regulates aldose reductase, a key enzyme in the polyol pathway. We evaluated whether soluble uric acid regulates aldose reductase expression both in cultured hepatocytes (HepG2 cells) and in the liver of hyperuricemic rats and whether this stimulation is associated with endogenous fructose production and fat accumulation. Uric acid dose-dependently stimulated aldose reductase expression in the HepG2 cells, and this stimulation was associated with endogenous fructose production and triglyceride accumulation. This stimulatory mechanism was mediated by uric acid-induced oxidative stress and stimulation of the transcription factor nuclear factor of activated T cells 5 (NFAT5). Uric acid also amplified the effects of elevated glucose levels to stimulate hepatocyte triglyceride accumulation. Hyperuricemic rats exhibited elevated hepatic aldose reductase expression, endogenous fructose accumulation, and fat buildup that was significantly reduced by co-administration of the xanthine oxidase inhibitor allopurinol. These results suggest that uric acid generated during fructose metabolism may act as a positive feedback mechanism that stimulates endogenous fructose production by stimulating aldose reductase in the polyol pathway. Our findings suggest an amplifying mechanism whereby soft drinks rich in glucose and fructose can induce NAFLD.


Assuntos
Tecido Adiposo/metabolismo , Aldeído Redutase/metabolismo , Frutose/biossíntese , Hepatopatia Gordurosa não Alcoólica/metabolismo , Polímeros/metabolismo , Ácido Úrico/farmacologia , Animais , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Frutose/metabolismo , Células Hep G2 , Humanos , Masculino , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/patologia , Estresse Oxidativo/efeitos dos fármacos , Polímeros/análise , Ratos , Ratos Wistar , Células Tumorais Cultivadas , Ácido Úrico/metabolismo
5.
J Clin Invest ; 128(6): 2226-2238, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29533924

RESUMO

Increasing evidence suggests a role for excessive intake of fructose in the Western diet as a contributor to the current epidemics of metabolic syndrome and obesity. Hereditary fructose intolerance (HFI) is a difficult and potentially lethal orphan disease associated with impaired fructose metabolism. In HFI, the deficiency of aldolase B results in the accumulation of intracellular phosphorylated fructose, leading to phosphate sequestration and depletion, increased adenosine triphosphate (ATP) turnover, and a plethora of conditions that lead to clinical manifestations such as fatty liver, hyperuricemia, Fanconi syndrome, and severe hypoglycemia. Unfortunately, there is currently no treatment for HFI, and avoiding sugar and fructose has become challenging in our society. In this report, through use of genetically modified mice and pharmacological inhibitors, we demonstrate that the absence or inhibition of ketohexokinase (Khk), an enzyme upstream of aldolase B, is sufficient to prevent hypoglycemia and liver and intestinal injury associated with HFI. Herein we provide evidence for the first time to our knowledge of a potential therapeutic approach for HFI. Mechanistically, our studies suggest that it is the inhibition of the Khk C isoform, not the A isoform, that protects animals from HFI.


Assuntos
Frutoquinases/antagonistas & inibidores , Frutoquinases/metabolismo , Intolerância à Frutose/enzimologia , Animais , Frutoquinases/genética , Frutose/genética , Frutose/metabolismo , Intolerância à Frutose/tratamento farmacológico , Intolerância à Frutose/genética , Frutose-Bifosfato Aldolase/antagonistas & inibidores , Frutose-Bifosfato Aldolase/genética , Frutose-Bifosfato Aldolase/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Camundongos , Camundongos Knockout
6.
PLoS One ; 11(6): e0157458, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27322374

RESUMO

OBJECTIVE: In developed countries with westernized diets, the excessive consumption of added sugar in beverages and highly refined and processed foods is associated with increased risk for obesity, diabetes, and cardiovascular diseases. As a major constituent of added sugars, fructose has been shown to cause a variety of adverse metabolic effects, such as impaired insulin sensitivity, hypertriglyceridemia, and oxidative stress. Recent studies have shown that ketohexokinase isoform C is the key enzyme responsible in fructose metabolism that drive's fructose's adverse effects. The objective of this study was to identify botanical ingredients with potential for inhibitory activity against ketohexokinase-C and fructose-induced metabolic effects by using a series of in vitro model systems. METHODS: Extracts from 406 botanicals and 1200 purified phytochemicals were screened (initial concentration of 50 µg/mL and 50 µM, respectively) for their inhibitory activity using a cell free, recombinant human ketohexokinase-C assay. Dose response evaluations were conducted on botanical extracts and phytochemicals that inhibited ketohexokinase-C by > 30% and > 40%, respectively. Two different extract lots of the top botanical candidates were further evaluated in lysates of HepG2 cells overexpressing ketohexokinase-C for inhibition of fructose-induced ATP depletion. In addition, extracts were evaluated in intact Hep G2 cells for inhibition of fructose-induced elevation of triglyceride and uric acid production. RESULTS: Among the botanical extracts, phloretin (Malus domestica) extracts were the most potent (IC50: 8.9-9.2 µg/mL) followed by extracts of Angelica archangelica (IC50: 22.6 µg/mL-57.3 µg/mL). Among the purified phytochemicals, methoxy-isobavachalcone (Psoralea corylifolia, IC50 = 0.2 µM) exhibited the highest potency against ketohexokinase isoform C activity followed by osthole (Angelica archangelica, IC50 = 0.7 µM), cratoxyarborenone E (Cratoxylum prunifolium, IC50 = 1.0 µM), and α-/γ-mangostin (Cratoxylum prunifolium, IC50 = 1.5 µM). Extracts of Angelica archangelica, Garcinia mangostana, Petroselinum crispum, and Scutellaria baicalensis exhibited ketohexokinase inhibitory activity and blocked fructose-induced ATP depletion and fructose-induced elevation in triglyerides and uric acid. CONCLUSIONS: Angelica archangelica, Garcinia mangostana, Petroselinum crispum, and Scutellaria baicalensis were the top four botanical candidiates identified with inhibitory activity against ketohexokinase-C. Future studies are needed to show proof of mechanism and the efficacy of these botanical extracts in humans to blunt the negative metabolic effects of fructose-containing added sugars.


Assuntos
Inibidores Enzimáticos/química , Frutoquinases/química , Frutose/metabolismo , Hipertrigliceridemia/tratamento farmacológico , Compostos Fitoquímicos/química , Angelica archangelica/química , Inibidores Enzimáticos/administração & dosagem , Frutoquinases/antagonistas & inibidores , Frutose/química , Garcinia mangostana/química , Células Hep G2 , Humanos , Hipertrigliceridemia/metabolismo , Insulina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Petroselinum/química , Compostos Fitoquímicos/administração & dosagem , Extratos Vegetais/administração & dosagem , Extratos Vegetais/química
7.
J Am Soc Nephrol ; 25(11): 2526-38, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24876114

RESUMO

Diabetes is associated with activation of the polyol pathway, in which glucose is converted to sorbitol by aldose reductase. Previous studies focused on the role of sorbitol in mediating diabetic complications. However, in the proximal tubule, sorbitol can be converted to fructose, which is then metabolized largely by fructokinase, also known as ketohexokinase, leading to ATP depletion, proinflammatory cytokine expression, and oxidative stress. We and others recently identified a potential deleterious role of dietary fructose in the generation of tubulointerstitial injury and the acceleration of CKD. In this study, we investigated the potential role of endogenous fructose production, as opposed to dietary fructose, and its metabolism through fructokinase in the development of diabetic nephropathy. Wild-type mice with streptozotocin-induced diabetes developed proteinuria, reduced GFR, and renal glomerular and proximal tubular injury. Increased renal expression of aldose reductase; elevated levels of renal sorbitol, fructose, and uric acid; and low levels of ATP confirmed activation of the fructokinase pathway. Furthermore, renal expression of inflammatory cytokines with macrophage infiltration was prominent. In contrast, diabetic fructokinase-deficient mice demonstrated significantly less proteinuria, renal dysfunction, renal injury, and inflammation. These studies identify fructokinase as a novel mediator of diabetic nephropathy and document a novel role for endogenous fructose production, or fructoneogenesis, in driving renal disease.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Frutoquinases/metabolismo , Frutose/biossíntese , Frutose/metabolismo , Túbulos Renais Proximais/enzimologia , Animais , Glicemia/metabolismo , Peso Corporal , Linhagem Celular Transformada , Quimiocinas/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/patologia , Humanos , Córtex Renal/enzimologia , Córtex Renal/patologia , Glomérulos Renais/citologia , Glomérulos Renais/patologia , Túbulos Renais Proximais/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polímeros/metabolismo
8.
FASEB J ; 28(8): 3339-50, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24755741

RESUMO

Reduced AMP kinase (AMPK) activity has been shown to play a key deleterious role in increased hepatic gluconeogenesis in diabetes, but the mechanism whereby this occurs remains unclear. In this article, we document that another AMP-dependent enzyme, AMP deaminase (AMPD) is activated in the liver of diabetic mice, which parallels with a significant reduction in AMPK activity and a significant increase in intracellular glucose accumulation in human HepG2 cells. AMPD activation is induced by a reduction in intracellular phosphate levels, which is characteristic of insulin resistance and diabetic states. Increased gluconeogenesis is mediated by reduced TORC2 phosphorylation at Ser171 by AMPK in these cells, as well as by the up-regulation of the rate-limiting enzymes PEPCK and G6Pc. The mechanism whereby AMPD controls AMPK activation depends on the production of a specific AMP downstream metabolite through AMPD, uric acid. In this regard, humans have higher uric acid levels than most mammals due to a mutation in uricase, the enzyme involved in uric acid degradation in most mammals, that developed during a period of famine in Europe 1.5 × 10(7) yr ago. Here, working with resurrected ancestral uricases obtained from early hominids, we show that their expression on HepG2 cells is enough to blunt gluconeogenesis in parallel with an up-regulation of AMPK activity. These studies identify a key role AMPD and uric acid in mediating hepatic gluconeogenesis in the diabetic state, via a mechanism involving AMPK down-regulation and overexpression of PEPCK and G6Pc. The uricase mutation in the Miocene likely provided a survival advantage to help maintain glucose levels under conditions of near starvation, but today likely has a role in the pathogenesis of diabetes.


Assuntos
AMP Desaminase/fisiologia , Gluconeogênese/fisiologia , Fígado/metabolismo , Inanição/fisiopatologia , Ácido Úrico/metabolismo , AMP Desaminase/antagonistas & inibidores , AMP Desaminase/genética , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Diabetes Mellitus Experimental/metabolismo , Europa (Continente) , Regulação Enzimológica da Expressão Gênica , Gluconeogênese/efeitos dos fármacos , Glucose-6-Fosfatase/biossíntese , Células Hep G2 , História Antiga , Hominidae/fisiologia , Humanos , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Fígado/enzimologia , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Complexos Multiproteicos/fisiologia , Fosfatos/metabolismo , Fosfatos/farmacologia , Fosfoenolpiruvato Carboxiquinase (ATP)/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Seleção Genética , Organismos Livres de Patógenos Específicos , Inanição/história , Serina-Treonina Quinases TOR/fisiologia , Transdução Genética , Urato Oxidase/genética , Urato Oxidase/história , Urato Oxidase/metabolismo , Ácido Úrico/farmacologia
9.
Proc Natl Acad Sci U S A ; 111(10): 3763-8, 2014 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-24550457

RESUMO

Uricase is an enzyme involved in purine catabolism and is found in all three domains of life. Curiously, uricase is not functional in some organisms despite its role in converting highly insoluble uric acid into 5-hydroxyisourate. Of particular interest is the observation that apes, including humans, cannot oxidize uric acid, and it appears that multiple, independent evolutionary events led to the silencing or pseudogenization of the uricase gene in ancestral apes. Various arguments have been made to suggest why natural selection would allow the accumulation of uric acid despite the physiological consequences of crystallized monosodium urate acutely causing liver/kidney damage or chronically causing gout. We have applied evolutionary models to understand the history of primate uricases by resurrecting ancestral mammalian intermediates before the pseudogenization events of this gene family. Resurrected proteins reveal that ancestral uricases have steadily decreased in activity since the last common ancestor of mammals gave rise to descendent primate lineages. We were also able to determine the 3D distribution of amino acid replacements as they accumulated during evolutionary history by crystallizing a mammalian uricase protein. Further, ancient and modern uricases were stably transfected into HepG2 liver cells to test one hypothesis that uricase pseudogenization allowed ancient frugivorous apes to rapidly convert fructose into fat. Finally, pharmacokinetics of an ancient uricase injected in rodents suggest that our integrated approach provides the foundation for an evolutionarily-engineered enzyme capable of treating gout and preventing tumor lysis syndrome in human patients.


Assuntos
Adaptação Biológica/genética , Evolução Molecular , Hominidae/genética , Modelos Moleculares , Filogenia , Conformação Proteica , Urato Oxidase/genética , Tecido Adiposo/metabolismo , Animais , Teorema de Bayes , Biologia Computacional , Primers do DNA/genética , Frutas/metabolismo , Células Hep G2 , Humanos , Modelos Biológicos , Modelos Genéticos , Pseudogenes/genética , Ratos , Ratos Sprague-Dawley , Urato Oxidase/química , Urato Oxidase/metabolismo
10.
Nat Commun ; 4: 2434, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24022321

RESUMO

Carbohydrates with high glycaemic index are proposed to promote the development of obesity, insulin resistance and fatty liver, but the mechanism by which this occurs remains unknown. High serum glucose concentrations are known to induce the polyol pathway and increase fructose generation in the liver. Here we show that this hepatic, endogenously produced fructose causes systemic metabolic changes. We demonstrate that mice unable to metabolize fructose are protected from an increase in energy intake and body weight, visceral obesity, fatty liver, elevated insulin levels and hyperleptinaemia after exposure to 10% glucose for 14 weeks. In normal mice, glucose consumption is accompanied by aldose reductase and polyol pathway activation in steatotic areas. In this regard, we show that aldose reductase-deficient mice are protected against glucose-induced fatty liver. We conclude that endogenous fructose generation and metabolism in the liver represents an important mechanism by which glucose promotes the development of metabolic syndrome.


Assuntos
Frutose/biossíntese , Frutose/metabolismo , Fígado/metabolismo , Fígado/patologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Aldeído Redutase/metabolismo , Animais , Metabolismo Energético , Fígado Gorduroso/metabolismo , Comportamento Alimentar , Frutoquinases/deficiência , Frutoquinases/metabolismo , Glucose/metabolismo , Células Hep G2 , Humanos , Fígado/enzimologia , Fígado/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polímeros/metabolismo
11.
Hepatology ; 58(5): 1632-43, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23813872

RESUMO

UNLABELLED: Fructose intake from added sugars has been implicated as a cause of nonalcoholic fatty liver disease. Here we tested the hypothesis that fructose may interact with a high-fat diet to induce fatty liver, and to determine if this was dependent on a key enzyme in fructose metabolism, fructokinase. Wild-type or fructokinase knockout mice were fed a low-fat (11%), high-fat (36%), or high-fat (36%) and high-sucrose (30%) diet for 15 weeks. Both wild-type and fructokinase knockout mice developed obesity with mild hepatic steatosis and no evidence of hepatic inflammation on a high-fat diet compared to a low-fat diet. In contrast, wild-type mice fed a high-fat and high-sucrose diet developed more severe hepatic steatosis with low-grade inflammation and fibrosis, as noted by increased CD68, tumor necrosis factor alpha, monocyte chemoattractant protein-1, alpha-smooth muscle actin, and collagen I and TIMP1 expression. These changes were prevented in the fructokinase knockout mice. CONCLUSION: An additive effect of high-fat and high-sucrose diet on the development of hepatic steatosis exists. Further, the combination of sucrose with high-fat diet may induce steatohepatitis. The protection in fructokinase knockout mice suggests a key role for fructose (from sucrose) in this development of steatohepatitis. These studies emphasize the important role of fructose in the development of fatty liver and nonalcoholic steatohepatitis.


Assuntos
Dieta Hiperlipídica , Fígado Gorduroso/etiologia , Frutoquinases/fisiologia , Sacarose/administração & dosagem , Animais , Ingestão de Energia , Frutose/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Aumento de Peso
12.
PLoS One ; 7(10): e47948, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23112875

RESUMO

Excessive dietary fructose intake may have an important role in the current epidemics of fatty liver, obesity and diabetes as its intake parallels the development of these syndromes and because it can induce features of metabolic syndrome. The effects of fructose to induce fatty liver, hypertriglyceridemia and insulin resistance, however, vary dramatically among individuals. The first step in fructose metabolism is mediated by fructokinase (KHK), which phosphorylates fructose to fructose-1-phosphate; intracellular uric acid is also generated as a consequence of the transient ATP depletion that occurs during this reaction. Here we show in human hepatocytes that uric acid up-regulates KHK expression thus leading to the amplification of the lipogenic effects of fructose. Inhibition of uric acid production markedly blocked fructose-induced triglyceride accumulation in hepatocytes in vitro and in vivo. The mechanism whereby uric acid stimulates KHK expression involves the activation of the transcription factor ChREBP, which, in turn, results in the transcriptional activation of KHK by binding to a specific sequence within its promoter. Since subjects sensitive to fructose often develop phenotypes associated with hyperuricemia, uric acid may be an underlying factor in sensitizing hepatocytes to fructose metabolism during the development of fatty liver.


Assuntos
Fígado Gorduroso/metabolismo , Frutoquinases/metabolismo , Frutose/metabolismo , Hepatócitos/metabolismo , Ácido Úrico/metabolismo , Alopurinol/farmacologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Inibidores Enzimáticos/farmacologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Frutoquinases/genética , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Ativação Transcricional , Regulação para Cima/efeitos dos fármacos , Ácido Úrico/antagonistas & inibidores
13.
PLoS One ; 7(11): e48801, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23152807

RESUMO

Fatty liver (hepatic steatosis) is associated with nucleotide turnover, loss of ATP and generation of adenosine monophosphate (AMP). It is well known that in fatty liver, activity of the AMP-activated kinase (AMPK) is reduced and that its stimulation can prevent hepatic steatosis by both enhancing fat oxidation and reducing lipogenesis. Here we show that another AMP dependent enzyme, AMPD2, has opposing effects on fatty acid oxidation when compared to AMPK. In human hepatocytres, AMPD2 activation -either by overexpression or by lowering intracellular phosphate levels with fructose- is associated with a significant reduction in AMPK activity. Likewise, silencing of AMPK spontaneously increases AMPD activity, demonstrating that these enzymes counter-regulate each other. Furthermore, we show that a downstream product of AMP metabolism through AMPD2, uric acid, can inhibit AMPK activity in human hepatocytes. Finally, we show that fructose-induced fat accumulation in hepatocytes is due to a dominant stimulation of AMPD2 despite stimulating AMPK. In this regard, AMPD2-deficient hepatocytes demonstrate a further activation of AMPK after fructose exposure in association with increased fatty acid oxidation, and conversely silencing AMPK enhances AMPD-dependent fat accumulation. In vivo, we show that sucrose fed rats also develop fatty liver that is blocked by metformin in association with both a reduction in AMPD activity and an increase in AMPK activity. In summary, AMPD and AMPK are both important in hepatic fat accumulation and counter-regulate each other. We present the novel finding that uric acid inhibits AMPK kinase activity in fructose-fed hepatocytes thus providing new insights into the pathogenesis of fatty liver.


Assuntos
AMP Desaminase/metabolismo , Adenilato Quinase/metabolismo , Fígado Gorduroso/metabolismo , Adenilato Quinase/genética , Animais , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Ativação Enzimática/efeitos dos fármacos , Gorduras/metabolismo , Fígado Gorduroso/enzimologia , Frutose/metabolismo , Frutose/farmacologia , Regulação da Expressão Gênica , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Isoenzimas , Masculino , Metformina/farmacologia , Oxirredução , Ratos , Ácido Úrico/metabolismo
14.
J Biol Chem ; 287(48): 40732-44, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23035112

RESUMO

BACKGROUND: Uric acid is an independent risk factor in fructose-induced fatty liver, but whether it is a marker or a cause remains unknown. RESULTS: Hepatocytes exposed to uric acid developed mitochondrial dysfunction and increased de novo lipogenesis, and its blockade prevented fructose-induced lipogenesis. CONCLUSION: Rather than a consequence, uric acid induces fatty liver SIGNIFICANCE: Hyperuricemic people are more prone to develop fructose-induced fatty liver. Metabolic syndrome represents a collection of abnormalities that includes fatty liver, and it currently affects one-third of the United States population and has become a major health concern worldwide. Fructose intake, primarily from added sugars in soft drinks, can induce fatty liver in animals and is epidemiologically associated with nonalcoholic fatty liver disease in humans. Fructose is considered lipogenic due to its ability to generate triglycerides as a direct consequence of the metabolism of the fructose molecule. Here, we show that fructose also stimulates triglyceride synthesis via a purine-degrading pathway that is triggered from the rapid phosphorylation of fructose by fructokinase. Generated AMP enters into the purine degradation pathway through the activation of AMP deaminase resulting in uric acid production and the generation of mitochondrial oxidants. Mitochondrial oxidative stress results in the inhibition of aconitase in the Krebs cycle, resulting in the accumulation of citrate and the stimulation of ATP citrate lyase and fatty-acid synthase leading to de novo lipogeneis. These studies provide new insights into the pathogenesis of hepatic fat accumulation under normal and diseased states.


Assuntos
Fígado Gorduroso/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Ácido Úrico/metabolismo , Frutose/metabolismo , Células Hep G2 , Humanos , Lipogênese , Triglicerídeos/metabolismo , Ácido Úrico/efeitos adversos
15.
Metabolism ; 60(9): 1259-70, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21489572

RESUMO

Fructose induces metabolic syndrome in rats; but studies have been criticized for using high concentrations of fructose that are not physiologic, for using only pure fructose, and for not controlling for energy intake. We tested the hypothesis that a 40% sucrose diet (containing 20% fructose) might induce features of metabolic syndrome in male breeder rats independent of excess energy intake. Male Sprague-Dawley breeder rats were pair fed 40% sucrose or isocaloric starch diet for 4 months and evaluated for metabolic syndrome and diabetes. In vitro studies were performed in rat insulinoma cells (RIN-m5F) exposed to uric acid, and markers of inflammation were assessed. Rats fed a 40% sucrose diet developed accelerated features of metabolic syndrome with up-regulation of fructose-dependent transporter Glut5 and fructokinase. Fatty liver and low-grade pancreatic inflammation also occurred. Uric acid was found to stimulate inflammatory mediators and oxidative stress in islet cells in vitro. Sucrose, at concentrations ingested by a subset of Americans, can accelerate metabolic syndrome, fatty liver, and type 2 diabetes mellitus in male breeder rats; and the effects are independent of excess energy intake.


Assuntos
Ingestão de Energia , Fígado Gorduroso/etiologia , Pancreatite/etiologia , Sacarose/toxicidade , Animais , Proteínas de Transporte de Ânions/genética , Linhagem Celular Tumoral , Frutose/metabolismo , Masculino , Síndrome Metabólica/etiologia , Ratos , Ratos Sprague-Dawley , Amido/administração & dosagem , Sacarose/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA