Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Nutrients ; 15(20)2023 Oct 16.
Article En | MEDLINE | ID: mdl-37892451

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.


Digestive System Diseases , Fructose Intolerance , Liver Diseases , Humans , Animals , Mice , Fructose Intolerance/genetics , Fructose Intolerance/diagnosis , Fructose/metabolism , Fructose-Bisphosphate Aldolase/genetics , Glucose/therapeutic use , Mice, Knockout
2.
Biomolecules ; 13(5)2023 04 30.
Article En | MEDLINE | ID: mdl-37238651

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.


High Fructose Corn Syrup , Kidney Diseases , Metabolic Syndrome , Mice , Animals , Metabolic Syndrome/complications , High Fructose Corn Syrup/adverse effects , Mice, Obese , Dietary Sucrose/adverse effects , Dietary Sucrose/metabolism , Obesity/etiology , Fructose/metabolism , Kidney Diseases/chemically induced , Fructokinases
3.
iScience ; 26(4): 106355, 2023 Apr 21.
Article En | MEDLINE | ID: mdl-36994079

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.

4.
iScience ; 25(8): 104694, 2022 Aug 19.
Article En | MEDLINE | ID: mdl-35847557

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.

5.
Nat Metab ; 3(9): 1189-1201, 2021 09.
Article En | MEDLINE | ID: mdl-34552272

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.


Metabolic Syndrome/metabolism , Nucleotides/metabolism , Obesity/metabolism , Taste , Uric Acid/metabolism , Animals , Energy Intake/drug effects , Metabolic Syndrome/chemically induced , Mice , Sodium Glutamate/pharmacology
6.
JCI Insight ; 6(1)2021 01 11.
Article En | MEDLINE | ID: mdl-33320834

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.


Fructose/metabolism , Metabolic Syndrome/metabolism , Receptors, Vasopressin/metabolism , Vasopressins/metabolism , Animals , Disease Models, Animal , Drinking/physiology , Fructokinases/metabolism , Fructose/administration & dosage , Hep G2 Cells , Humans , Liver/metabolism , Male , Metabolic Syndrome/chemically induced , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Vasopressin/deficiency , Receptors, Vasopressin/genetics , Vasopressins/antagonists & inhibitors , Vasopressins/biosynthesis
7.
Am J Physiol Endocrinol Metab ; 319(2): E276-E290, 2020 08 01.
Article En | MEDLINE | ID: mdl-32574112

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.


Dietary Sucrose/adverse effects , Metabolic Syndrome/etiology , Obesity/etiology , Taste/physiology , Animals , Dietary Sucrose/administration & dosage , Food Preferences/physiology , Fructose/administration & dosage , Fructose/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Purinergic P2X2/deficiency , Receptors, Purinergic P2X2/physiology , Receptors, Purinergic P2X3/deficiency , Receptors, Purinergic P2X3/physiology
8.
Am J Physiol Renal Physiol ; 317(4): F941-F948, 2019 10 01.
Article En | MEDLINE | ID: mdl-31411075

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.


Energy Metabolism , Kidney/metabolism , Mitochondria/metabolism , Obesity/complications , Obesity/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Adenine/toxicity , Adenosine Triphosphate/metabolism , Animals , Kidney Function Tests , Kidney Tubules/pathology , Mice , Mice, Inbred C57BL , Mice, Obese , Sarcopenia/etiology , Sarcopenia/metabolism
9.
J Biol Chem ; 294(11): 4272-4281, 2019 03 15.
Article En | MEDLINE | ID: mdl-30651350

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.


Adipose Tissue/metabolism , Aldehyde Reductase/metabolism , Fructose/biosynthesis , Non-alcoholic Fatty Liver Disease/metabolism , Polymers/metabolism , Uric Acid/pharmacology , Animals , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fructose/metabolism , Hep G2 Cells , Humans , Male , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Polymers/analysis , Rats , Rats, Wistar , Tumor Cells, Cultured , Uric Acid/metabolism
11.
Proc Natl Acad Sci U S A ; 115(12): 3138-3143, 2018 03 20.
Article En | MEDLINE | ID: mdl-29507217

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.


Fructose/metabolism , Leptin/blood , Non-alcoholic Fatty Liver Disease/chemically induced , Obesity/chemically induced , Sodium Chloride, Dietary/adverse effects , Adult , Aged , Aged, 80 and over , Animals , Diabetes Mellitus/chemically induced , Fructokinases/genetics , Humans , Leptin/genetics , Metabolic Syndrome/chemically induced , Metabolic Syndrome/genetics , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/metabolism , Sucrose/adverse effects , Sucrose/analogs & derivatives , Transcription Factors/genetics , Transcription Factors/metabolism
12.
J Clin Invest ; 128(6): 2226-2238, 2018 06 01.
Article En | MEDLINE | ID: mdl-29533924

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.


Fructokinases/antagonists & inhibitors , Fructokinases/metabolism , Fructose Intolerance/enzymology , Animals , Fructokinases/genetics , Fructose/genetics , Fructose/metabolism , Fructose Intolerance/drug therapy , Fructose Intolerance/genetics , Fructose-Bisphosphate Aldolase/antagonists & inhibitors , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Mice , Mice, Knockout
13.
Nat Commun ; 8: 14181, 2017 02 13.
Article En | MEDLINE | ID: mdl-28194018

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.


Acute Kidney Injury/prevention & control , Fructokinases/antagonists & inhibitors , Kidney/drug effects , Luteolin/pharmacology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Aldehyde Reductase/metabolism , Animals , Cell Line , Fructokinases/genetics , Fructokinases/metabolism , Fructose/metabolism , Fructose/urine , Humans , Ischemia/complications , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Protective Agents/pharmacology , Uric Acid/metabolism
14.
PLoS One ; 12(1): e0169137, 2017.
Article En | MEDLINE | ID: mdl-28081152

BACKGROUND: Epidemics of chronic kidney disease (CKD) not due to diabetes mellitus (DM) or hypertension have been observed among individuals working in hot environments in several areas of the world. Experimental models have documented that recurrent heat stress and water restriction can lead to CKD, and the mechanism may be mediated by hyperosmolarity that activates pathways (vasopressin, aldose reductase-fructokinase) that induce renal injury. Here we tested the hypothesis that elevated serum sodium, which reflects serum osmolality, may be an independent risk factor for the development of CKD. METHODS: This study was a large-scale, single-center, retrospective 5-year cohort study at Center for Preventive Medicine, St. Luke's International Hospital, Tokyo, Japan, between 2004 and 2009. We analyzed 13,201 subjects who underwent annual medical examination of which 12,041 subjects (age 35 to 85) without DM and/or CKD were enrolled. This analysis evaluated age, sex, body mass index, abdominal circumference, hypertension, dyslipidemia, hyperuricemia, fasting glucose, BUN, serum sodium, potassium, chloride and calculated serum osmolarity. RESULTS: Elevated serum sodium was an independent risk factor for development of CKD (OR: 1.03, 95% CI, 1.00-1.07) after adjusted regression analysis with an 18 percent increased risk for every 5 mmol/L change in serum sodium. Calculated serum osmolarity was also an independent risk factor for CKD (OR: 1.04; 95% CI, 1.03-1.05) as was BUN (OR: 1.08; 95% CI, 1.06-1.10) (independent of serum creatinine). CONCLUSIONS: Elevated serum sodium and calculated serum osmolarity are independent risk factors for developing CKD. This finding supports the role of limiting salt intake and preventing dehydration to reduce risk of CKD.


Renal Insufficiency, Chronic , Sodium/blood , Urinary Calculi , Adolescent , Adult , Cross-Sectional Studies , Female , Humans , Japan , Male , Middle Aged , Osmolar Concentration , Prevalence , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/etiology , Retrospective Studies , Risk Factors , Sex Factors , Urinary Calculi/blood , Urinary Calculi/complications , Urinary Calculi/epidemiology
15.
Int J Cardiol ; 223: 500-506, 2016 Nov 15.
Article En | MEDLINE | ID: mdl-27552564

BACKGROUND: Cardiometabolic disruptions such as insulin resistance, obesity, high blood pressure, hyperglycemia, and dyslipidemias, are known to increase the risk for cardiovascular and metabolic diseases such as type 2 diabetes mellitus and atherosclerosis. Several screening tools for assessing cardiometabolic risk have been developed including the TG/HDLc ratio, which has been, demonstrated to possess a strong association with insulin resistance and coronary disease. Dietary modifications, together with regular moderate exercise have proven to be effective in attenuating cardiometabolic disruptions. However, they often exhibit poor long-term patient compliance. Nutraceutics, including (-)-epicatechin (EPI), have gained increasing interest as coadjuvant effective and safe therapies that are able to attenuate hypertension, hyperglycemia, hyperinsulinemia, hypertriglyceridemia and hypoalphalipoproteinemia. METHODS: The aims of this study were: 1) to compare the in vitro effect of EPI vs. (+)-catechin on fructose induced triglyceride accumulation and mitochondrial function in Hep2 cells in culture, 2) to evaluate the efficacy of EPI treatment in reducing fasting blood triglycerides and improving the TG/HDLc ratio in hypertriglyceridemic patients with a total daily dose of 100mg of EPI. Secondary clinical variables included total cholesterol, LDLc, fructosamine, glucose, insulin, and high sensitivity C-reactive protein blood levels. RESULTS AND CONCLUSION: Our results provide preliminary evidence as to favorable effects of EPI on glycemia homeostasis, lipid profile and systemic inflammation such bioactive actions are not class-effects (i.e. limited to their antioxidant potential) but instead, may result from the specific activation of associated downstream signaling pathways since catechin has no effects.


Cardiovascular Diseases/etiology , Catechin/therapeutic use , Cholesterol, HDL/blood , Hypertriglyceridemia/drug therapy , Metabolic Syndrome/etiology , Triglycerides/blood , Adolescent , Adult , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Cells, Cultured , Double-Blind Method , Female , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Male , Metabolic Syndrome/blood , Metabolic Syndrome/drug therapy , Middle Aged , Risk Factors , Young Adult
16.
PLoS One ; 11(6): e0157458, 2016.
Article En | MEDLINE | ID: mdl-27322374

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.


Enzyme Inhibitors/chemistry , Fructokinases/chemistry , Fructose/metabolism , Hypertriglyceridemia/drug therapy , Phytochemicals/chemistry , Angelica archangelica/chemistry , Enzyme Inhibitors/administration & dosage , Fructokinases/antagonists & inhibitors , Fructose/chemistry , Garcinia mangostana/chemistry , Hep G2 Cells , Humans , Hypertriglyceridemia/metabolism , Insulin/metabolism , Oxidative Stress/drug effects , Petroselinum/chemistry , Phytochemicals/administration & dosage , Plant Extracts/administration & dosage , Plant Extracts/chemistry
17.
PLoS One ; 10(4): e0123509, 2015.
Article En | MEDLINE | ID: mdl-25856396

Hibernating animals develop fatty liver when active in summertime and undergo a switch to a fat oxidation state in the winter. We hypothesized that this switch might be determined by AMP and the dominance of opposing effects: metabolism through AMP deaminase (AMPD2) (summer) and activation of AMP-activated protein kinase (AMPK) (winter). Liver samples were obtained from 13-lined ground squirrels at different times during the year, including summer and multiples stages of winter hibernation, and fat synthesis and ß-fatty acid oxidation were evaluated. Changes in fat metabolism were correlated with changes in AMPD2 activity and intrahepatic uric acid (downstream product of AMPD2), as well as changes in AMPK and intrahepatic ß-hydroxybutyrate (a marker of fat oxidation). Hepatic fat accumulation occurred during the summer with relatively increased enzymes associated with fat synthesis (FAS, ACL and ACC) and decreased enoyl CoA hydratase (ECH1) and carnitine palmitoyltransferase 1A (CPT1A), rate limiting enzymes of fat oxidation. In summer, AMPD2 activity and intrahepatic uric acid levels were high and hepatic AMPK activity was low. In contrast, the active phosphorylated form of AMPK and ß-hydroxybutyrate both increased during winter hibernation. Therefore, changes in AMPD2 and AMPK activity were paralleled with changes in fat synthesis and fat oxidation rates during the summer-winter cycle. These data illuminate the opposing forces of metabolism of AMP by AMPD2 and its availability to activate AMPK as a switch that governs fat metabolism in the liver of hibernating ground squirrel.


AMP Deaminase/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/metabolism , Hibernation/physiology , Animals , Fatty Acids/metabolism , Glucose/metabolism , Hibernation/genetics , Lipid Metabolism/genetics , Liver/metabolism , Liver/physiology , Oxidation-Reduction , Sciuridae/metabolism , Sciuridae/physiology , Seasons
18.
J Am Soc Nephrol ; 25(11): 2526-38, 2014 Nov.
Article En | MEDLINE | ID: mdl-24876114

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.


Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Fructokinases/metabolism , Fructose/biosynthesis , Fructose/metabolism , Kidney Tubules, Proximal/enzymology , Animals , Blood Glucose/metabolism , Body Weight , Cell Line, Transformed , Chemokines/metabolism , Cytokines/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Humans , Kidney Cortex/enzymology , Kidney Cortex/pathology , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/pathology , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Polymers/metabolism
19.
FASEB J ; 28(8): 3339-50, 2014 Aug.
Article En | MEDLINE | ID: mdl-24755741

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.


AMP Deaminase/physiology , Gluconeogenesis/physiology , Liver/metabolism , Starvation/physiopathology , Uric Acid/metabolism , AMP Deaminase/antagonists & inhibitors , AMP Deaminase/genetics , AMP-Activated Protein Kinases/physiology , Animals , Diabetes Mellitus, Experimental/metabolism , Europe , Gene Expression Regulation, Enzymologic , Gluconeogenesis/drug effects , Glucose-6-Phosphatase/biosynthesis , Hep G2 Cells , History, Ancient , Hominidae/physiology , Humans , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Liver/enzymology , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Models, Biological , Multiprotein Complexes/physiology , Phosphates/metabolism , Phosphates/pharmacology , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , Recombinant Fusion Proteins/metabolism , Selection, Genetic , Specific Pathogen-Free Organisms , Starvation/history , TOR Serine-Threonine Kinases/physiology , Transduction, Genetic , Urate Oxidase/genetics , Urate Oxidase/history , Urate Oxidase/metabolism , Uric Acid/pharmacology
20.
Proc Natl Acad Sci U S A ; 111(10): 3763-8, 2014 Mar 11.
Article En | MEDLINE | ID: mdl-24550457

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.


Adaptation, Biological/genetics , Evolution, Molecular , Hominidae/genetics , Models, Molecular , Phylogeny , Protein Conformation , Urate Oxidase/genetics , Adipose Tissue/metabolism , Animals , Bayes Theorem , Computational Biology , DNA Primers/genetics , Fruit/metabolism , Hep G2 Cells , Humans , Models, Biological , Models, Genetic , Pseudogenes/genetics , Rats , Rats, Sprague-Dawley , Urate Oxidase/chemistry , Urate Oxidase/metabolism
...