Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 115(12): 3138-3143, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29507217

RESUMO

Dietary guidelines for obesity typically focus on three food groups (carbohydrates, fat, and protein) and caloric restriction. Intake of noncaloric nutrients, such as salt, are rarely discussed. However, recently high salt intake has been reported to predict the development of obesity and insulin resistance. The mechanism for this effect is unknown. Here we show that high intake of salt activates the aldose reductase-fructokinase pathway in the liver and hypothalamus, leading to endogenous fructose production with the development of leptin resistance and hyperphagia that cause obesity, insulin resistance, and fatty liver. A high-salt diet was also found to predict the development of diabetes and nonalcoholic fatty liver disease in a healthy population. These studies provide insights into the pathogenesis of obesity and diabetes and raise the potential for reduction in salt intake as an additional interventional approach for reducing the risk for developing obesity and metabolic syndrome.


Assuntos
Frutose/metabolismo , Leptina/sangue , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Obesidade/induzido quimicamente , Cloreto de Sódio na Dieta/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Diabetes Mellitus/induzido quimicamente , Frutoquinases/genética , Humanos , Leptina/genética , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/metabolismo , Sacarose/efeitos adversos , Sacarose/análogos & derivados , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
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
3.
Am J Physiol Endocrinol Metab ; 319(2): E276-E290, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32574112

RESUMO

Intake of sugars, especially the fructose component, is strongly associated with the development of obesity and metabolic syndrome, but the relative role of taste versus metabolism in driving preference, intake, and metabolic outcome is not fully understood. We aimed to evaluate the preference for sweet substances and the tendency to develop metabolic syndrome in response to these sugars in mice lacking functional taste signaling [P2X2 (P2X purinoreceptor 2)/P2X3 (P2X purinoreceptor 3) double knockout mice (DKO)] and mice unable to metabolize fructose (fructokinase knockout mice). Of interest, our data indicate that despite their inability to taste sweetness, P2X2/3 DKO mice still prefer caloric sugars (including fructose and glucose) to water in long-term testing, although with diminished preference compared with control mice. Despite reduced intake of caloric sugars by P2X2/3 DKO animals, the DKO mice still show increased levels of the sugar-dependent hormone FGF21 (fibroblast growth factor 21) in plasma and liver. Despite lower sugar intake, taste-blind mice develop severe features of metabolic syndrome due to reduced sensitivity to leptin, reduced ability to mobilize and oxidize fats, and increased hepatic de novo lipogenesis. In contrast to P2X2/3 DKO and wild-type mice, fructokinase knockout mice, which cannot metabolize fructose and are protected against fructose-induced metabolic syndrome, demonstrate reduced preference and intake for all fructose-containing sugars tested but not for glucose or artificial sweeteners. Based on these observations, we conclude that sugar can induce metabolic syndrome in mice independently of its sweet properties. Furthermore, our data demonstrate that the metabolism of fructose is necessary for sugar to drive intake and preference in mice.


Assuntos
Sacarose Alimentar/efeitos adversos , Síndrome Metabólica/etiologia , Obesidade/etiologia , Paladar/fisiologia , Animais , Sacarose Alimentar/administração & dosagem , Preferências Alimentares/fisiologia , Frutose/administração & dosagem , Frutose/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Purinérgicos P2X2/deficiência , Receptores Purinérgicos P2X2/fisiologia , Receptores Purinérgicos P2X3/deficiência , Receptores Purinérgicos P2X3/fisiologia
4.
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
5.
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
6.
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
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.
Kidney Int ; 86(2): 294-302, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24336030

RESUMO

The epidemic of chronic kidney disease in Nicaragua (Mesoamerican nephropathy) has been linked with recurrent dehydration. Here we tested whether recurrent dehydration may cause renal injury by activation of the polyol pathway, resulting in the generation of endogenous fructose in the kidney that might subsequently induce renal injury via metabolism by fructokinase. Wild-type and fructokinase-deficient mice were subjected to recurrent heat-induced dehydration. One group of each genotype was provided water throughout the day and the other group was hydrated at night, after the dehydration. Both groups received the same total hydration in 24 h. Wild-type mice that received delayed hydration developed renal injury, with elevated serum creatinine, increased urinary NGAL, proximal tubular injury, and renal inflammation and fibrosis. This was associated with activation of the polyol pathway, with increased renal cortical sorbitol and fructose levels. Fructokinase-knockout mice with delayed hydration were protected from renal injury. Thus, recurrent dehydration can induce renal injury via a fructokinase-dependent mechanism, likely from the generation of endogenous fructose via the polyol pathway. Access to sufficient water during the dehydration period can protect mice from developing renal injury. These studies provide a potential mechanism for Mesoamerican nephropathy.


Assuntos
Desidratação/enzimologia , Frutoquinases/metabolismo , Rim/lesões , Aldeído Redutase/metabolismo , Animais , Pressão Sanguínea , Desidratação/complicações , Modelos Animais de Doenças , Frutoquinases/deficiência , Frutoquinases/genética , Rim/patologia , Rim/fisiopatologia , Córtex Renal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Insuficiência Renal Crônica/enzimologia , Insuficiência Renal Crônica/etiologia , Ácido Úrico/metabolismo
9.
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
10.
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
11.
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.

12.
Nutrients ; 15(20)2023 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-37892451

RESUMO

Excessive intake of sugar, and particularly fructose, is closely associated with the development and progression of metabolic syndrome in humans and animal models. However, genetic disorders in fructose metabolism have very different consequences. While the deficiency of fructokinase, the first enzyme involved in fructose metabolism, is benign and somewhat desirable, missense mutations in the second enzyme, aldolase B, causes a very dramatic and sometimes lethal condition known as hereditary fructose intolerance (HFI). To date, there is no cure for HFI, and treatment is limited to avoiding fructose and sugar. Because of this, for subjects with HFI, glucose is their sole source of carbohydrates in the diet. However, clinical symptoms still occur, suggesting that either low amounts of fructose are still being consumed or, alternatively, fructose is being produced endogenously in the body. Here, we demonstrate that as a consequence of consuming high glycemic foods, the polyol pathway, a metabolic route in which fructose is produced from glucose, is activated, triggering a deleterious mechanism whereby glucose, sorbitol and alcohol induce severe liver disease and growth retardation in aldolase B knockout mice. We show that generically and pharmacologically blocking this pathway significantly improves metabolic dysfunction and thriving and increases the tolerance of aldolase B knockout mice to dietary triggers of endogenous fructose production.


Assuntos
Doenças do Sistema Digestório , Intolerância à Frutose , Hepatopatias , Humanos , Animais , Camundongos , Intolerância à Frutose/genética , Intolerância à Frutose/diagnóstico , Frutose/metabolismo , Frutose-Bifosfato Aldolase/genética , Glucose/uso terapêutico , Camundongos Knockout
13.
Biomolecules ; 13(5)2023 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-37238651

RESUMO

The presence of obesity and metabolic syndrome is strongly linked with chronic kidney disease (CKD), but the mechanisms responsible for the association are poorly understood. Here, we tested the hypothesis that mice with obesity and metabolic syndrome might have increased susceptibility to CKD from liquid high fructose corn syrup (HFCS) by favoring the absorption and utilization of fructose. We evaluated the pound mouse model of metabolic syndrome to determine if it showed baseline differences in fructose transport and metabolism and whether it was more susceptible to chronic kidney disease when administered HFCS. Pound mice have increased expression of fructose transporter (Glut5) and fructokinase (the limiting enzyme driving fructose metabolism) associated with enhanced fructose absorption. Pound mice receiving HFCS rapidly develop CKD with increased mortality rates associated with intrarenal mitochondria loss and oxidative stress. In pound mice lacking fructokinase, the effect of HFCS to cause CKD and early mortality was aborted, associated with reductions in oxidative stress and fewer mitochondria loss. Obesity and metabolic syndrome show increased susceptibility to fructose-containing sugars and increased risk for CKD and mortality. Lowering added sugar intake may be beneficial in reducing the risk for CKD in subjects with metabolic syndrome.


Assuntos
Xarope de Milho Rico em Frutose , Nefropatias , Síndrome Metabólica , Camundongos , Animais , Síndrome Metabólica/complicações , Xarope de Milho Rico em Frutose/efeitos adversos , Camundongos Obesos , Sacarose Alimentar/efeitos adversos , Sacarose Alimentar/metabolismo , Obesidade/etiologia , Frutose/metabolismo , Nefropatias/induzido quimicamente , Frutoquinases
14.
iScience ; 25(8): 104694, 2022 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-35847557

RESUMO

Steroid-sensitive nephrotic syndrome (SSNS) in childhood is usually due to minimal change disease (MCD). Unlike many glomerular conditions, SSNS/MCD is commonly precipitated by respiratory infections. Of interest, pulmonary inflammation releases surfactants in circulation which are soluble agonists of SIRPα, a podocyte receptor that regulates integrin signaling. Here, we characterized this pulmonary-renal connection in MCD and performed studies to determine its importance. Children with SSNS/MCD in relapse but not remission had elevated plasma surfactants and urinary SIRPα. Sera from relapsing subjects triggered podocyte SIRPα signaling via tyrosine phosphatase SHP-2 and nephrin dephosphorylation, a marker of podocyte activation. Further, addition of surfactants to MCD sera from patients in remission replicated these findings. Similarly, nasal instillation of toll-like receptor 3 and 4 agonists in mice resulted in elevated serum surfactants and their binding to glomeruli triggering proteinuria. Together, our data document a critical pulmonary-podocyte signaling pathway involving surfactants and SIRPα signaling in SSNS/MCD.

15.
J Biol Chem ; 285(41): 31694-703, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20639513

RESUMO

Expression of aquaporin-1 (AQP1) and -2 (AQP2) channels in the kidney are critical for the maintenance of water homeostasis and the operation of the urinary concentrating mechanism. Hypertonic stress induced in inner medullary (IMCD3) cells by addition of NaCl to the medium substantially up-regulated the mRNA and protein expression of AQP1, suggesting that its activation occurs at a transcriptional and a translational levels. In contrast, no up-regulation of AQP1 was observed when these cells were exposed to the same tonicity by addition of urea. To explore the transcriptional activation of aqp1 under hypertonic stress, we examined the role of the transcription factor associated with hypertonicity, TonEBP. Treatment of IMCD3 cells with the TonEBP inhibitor rottlerin or silencing its expression with specific shRNA technology led to a substantial reduction in AQP1 expression under hypertonic conditions. Moreover, we defined a conserved TonEBP binding site located 811 bp upstream of the aqp1 exon that is essential for its expression. Single site-directed mutation of this TonE site led to a 54 ± 5% (p < 0.01) decrease in AQP1 luciferase-driven activity under hypertonic stress. TonEBP mutant mice display marked decrement in the expression of AQP1 in the inner medulla. In conclusion, these data demonstrate that TonEBP is necessary for the regulation of AQP1 expression in the inner medulla of the kidney under hypertonic conditions.


Assuntos
Aquaporina 1/biossíntese , Medula Renal/metabolismo , Elementos de Resposta/fisiologia , Fatores de Transcrição/metabolismo , Regulação para Cima/fisiologia , Acetofenonas/farmacologia , Animais , Aquaporina 1/genética , Aquaporina 2/genética , Aquaporina 2/metabolismo , Benzopiranos/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Inativação Gênica , Camundongos , Camundongos Mutantes , Mutagênese Sítio-Dirigida , Mutação , Pressão Osmótica/efeitos dos fármacos , Pressão Osmótica/fisiologia , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , Cloreto de Sódio/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Regulação para Cima/efeitos dos fármacos , Ureia/farmacologia
16.
Nat Metab ; 3(9): 1189-1201, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34552272

RESUMO

Umami refers to the savoury taste that is mediated by monosodium glutamate (MSG) and enhanced by inosine monophosphate and other nucleotides. Umami foods have been suggested to increase the risk for obesity and metabolic syndrome but the mechanism is not understood. Here we show that MSG induces obesity, hypothalamic inflammation and central leptin resistance in male mice through the induction of AMP deaminase 2 and purine degradation. Mice lacking AMP deaminase 2 in both hepatocytes and neurons are protected from MSG-induced metabolic syndrome. This protection can be overcome by supplementation with inosine monophosphate, most probably owing to its degradation to uric acid as the effect can be blocked with allopurinol. Thus, umami foods induce obesity and metabolic syndrome by engaging the same purine nucleotide degradation pathway that is also activated by fructose and salt consumption. We suggest that the three tastes-sweet, salt and umami-developed to encourage food intake to facilitate energy storage and survival but drive obesity and diabetes in the setting of excess intake through similar mechanisms.


Assuntos
Síndrome Metabólica/metabolismo , Nucleotídeos/metabolismo , Obesidade/metabolismo , Paladar , Ácido Úrico/metabolismo , Animais , Ingestão de Energia/efeitos dos fármacos , Síndrome Metabólica/induzido quimicamente , Camundongos , Glutamato de Sódio/farmacologia
17.
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
18.
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
20.
Nat Commun ; 8: 14181, 2017 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-28194018

RESUMO

Acute kidney injury is associated with high mortality, especially in intensive care unit patients. The polyol pathway is a metabolic route able to convert glucose into fructose. Here we show the detrimental role of endogenous fructose production by the polyol pathway and its metabolism through fructokinase in the pathogenesis of ischaemic acute kidney injury (iAKI). Consistent with elevated urinary fructose in AKI patients, mice undergoing iAKI show significant polyol pathway activation in the kidney cortex characterized by high levels of aldose reductase, sorbitol and endogenous fructose. Wild type but not fructokinase knockout animals demonstrate severe kidney injury associated with ATP depletion, elevated uric acid, oxidative stress and inflammation. Interestingly, both the renal injury and dysfunction in wild-type mice undergoing iAKI is significantly ameliorated when exposed to luteolin, a recently discovered fructokinase inhibitor. This study demonstrates a role for fructokinase and endogenous fructose as mediators of acute renal disease.


Assuntos
Injúria Renal Aguda/prevenção & controle , Frutoquinases/antagonistas & inibidores , Rim/efeitos dos fármacos , Luteolina/farmacologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Aldeído Redutase/metabolismo , Animais , Linhagem Celular , Frutoquinases/genética , Frutoquinases/metabolismo , Frutose/metabolismo , Frutose/urina , Humanos , Isquemia/complicações , Rim/metabolismo , Rim/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Ácido Úrico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA