451
|
Affiliation(s)
- Claudio Borghi
- From the Department of Medical and Surgical Sciences, University of Bologna, Bologna Italy (C.B.); and Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy (G.D.)
| | - Giovambattista Desideri
- From the Department of Medical and Surgical Sciences, University of Bologna, Bologna Italy (C.B.); and Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy (G.D.)
| |
Collapse
|
452
|
Yamazaki M, Munetsuna E, Yamada H, Ando Y, Mizuno G, Murase Y, Kondo K, Ishikawa H, Teradaira R, Suzuki K, Ohashi K. Fructose consumption induces hypomethylation of hepatic mitochondrial DNA in rats. Life Sci 2016; 149:146-52. [PMID: 26869391 DOI: 10.1016/j.lfs.2016.02.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/11/2015] [Accepted: 02/06/2016] [Indexed: 10/22/2022]
Abstract
AIMS Fructose may play a crucial role in the pathogenesis of metabolic syndrome (MetS). However, the pathogenic mechanism of the fructose-induced MetS has not yet been investigated fully. Recently, several reports have investigated the association between mitochondrial DNA (mtDNA) and MetS. We examined the effect of fructose-rich diets on mtDNA content, transcription, and epigenetic changes. MAIN METHODS Four-week-old male Sprague-Dawley rats were offered a 20% fructose solution for 14weeks. We quantified mRNAs for hepatic mitochondrial genes and analyzed the mtDNA methylation (5-mC and 5-hmC) levels using ELISA kits. KEY FINDINGS Histological analysis revealed non-alcoholic fatty liver disease (NAFLD) in fructose-fed rats. Hepatic mtDNA content and transcription were higher in fructose-fed rats than in the control group. Global hypomethylation of mtDNA was also observed in fructose-fed rats. SIGNIFICANCE We showed that fructose consumption stimulates hepatic mtDNA-encoded gene expression. This phenomenon might be due to epigenetic changes in mtDNA. Fructose-induced mitochondrial epigenetic changes appear to be a novel mechanism underlying the pathology of MetS and NAFLD.
Collapse
Affiliation(s)
- Mirai Yamazaki
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hiroya Yamada
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake, Japan.
| | - Yoshitaka Ando
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Genki Mizuno
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Yuri Murase
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Kanako Kondo
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Hiroaki Ishikawa
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Ryoji Teradaira
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Koji Suzuki
- Department of Public Health, Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Koji Ohashi
- Department of Clinical Biochemistry, Fujita Health University School of Health Sciences, Toyoake, Japan.
| |
Collapse
|
453
|
Zhao L, Guo X, Wang O, Zhang H, Wang Y, Zhou F, Liu J, Ji B. Fructose and glucose combined with free fatty acids induce metabolic disorders in HepG2 cell: A new model to study the impacts of high-fructose/sucrose and high-fat diets in vitro. Mol Nutr Food Res 2016; 60:909-21. [DOI: 10.1002/mnfr.201500635] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/26/2015] [Accepted: 12/29/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Liang Zhao
- Beijing Key Laboratory of Functional Food from Plant Resources; College of Food Science and Nutritional Engineering; China Agricultural University; Beijing P. R. China
| | - Xiaoxuan Guo
- Beijing Key Laboratory of Functional Food from Plant Resources; College of Food Science and Nutritional Engineering; China Agricultural University; Beijing P. R. China
| | - Ou Wang
- Beijing Key Laboratory of Functional Food from Plant Resources; College of Food Science and Nutritional Engineering; China Agricultural University; Beijing P. R. China
| | | | - Yong Wang
- Beijing Key Laboratory of Functional Food from Plant Resources; College of Food Science and Nutritional Engineering; China Agricultural University; Beijing P. R. China
| | - Feng Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources; College of Food Science and Nutritional Engineering; China Agricultural University; Beijing P. R. China
| | - Jia Liu
- China National Research Institute of Food and Fermentation Industries; Beijing P. R. China
| | - Baoping Ji
- Beijing Key Laboratory of Functional Food from Plant Resources; College of Food Science and Nutritional Engineering; China Agricultural University; Beijing P. R. China
| |
Collapse
|
454
|
Saben JL, Asghar Z, Rhee JS, Drury A, Scheaffer S, Moley KH. Excess Maternal Fructose Consumption Increases Fetal Loss and Impairs Endometrial Decidualization in Mice. Endocrinology 2016; 157:956-68. [PMID: 26677880 PMCID: PMC4733112 DOI: 10.1210/en.2015-1618] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The most significant increase in metabolic syndrome over the previous decade occurred in women of reproductive age, which is alarming given that metabolic syndrome is associated with reproductive problems including subfertility and early pregnancy loss. Individuals with metabolic syndrome often consume excess fructose, and several studies have concluded that excess fructose intake contributes to metabolic syndrome development. Here, we examined the effects of increased fructose consumption on pregnancy outcomes in mice. Female mice fed a high-fructose diet (HFrD) for 6 weeks developed glucose intolerance and mild fatty liver but did not develop other prominent features of metabolic syndrome such as weight gain, hyperglycemia, and hyperinsulinemia. Upon mating, HFrD-exposed mice had lower pregnancy rates and smaller litters at midgestation than chow-fed controls. To explain this phenomenon, we performed artificial decidualization experiments and found that HFrD consumption impaired decidualization. This appeared to be due to decreased circulating progesterone as exogenous progesterone administration rescued decidualization. Furthermore, HFrD intake was associated with decreased bone morphogenetic protein 2 expression and signaling, both of which were restored by exogenous progesterone. Finally, expression of forkhead box O1 and superoxide dismutase 2 [Mn] proteins were decreased in the uteri of HFrD-fed mice, suggesting that HFrD consumption promotes a prooxidative environment in the endometrium. In summary, these data suggest that excess fructose consumption impairs murine fertility by decreasing steroid hormone synthesis and promoting an adverse uterine environment.
Collapse
Affiliation(s)
- Jessica L Saben
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Zeenat Asghar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Julie S Rhee
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Andrea Drury
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Suzanne Scheaffer
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
455
|
Fan N, Zhang L, Xia Z, Peng L, Wang Y, Peng Y. Sex-Specific Association between Serum Uric Acid and Nonalcoholic Fatty Liver Disease in Type 2 Diabetic Patients. J Diabetes Res 2016; 2016:3805372. [PMID: 27382573 PMCID: PMC4921134 DOI: 10.1155/2016/3805372] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/11/2016] [Accepted: 05/15/2016] [Indexed: 12/21/2022] Open
Abstract
Across-sectional study was performed in 541 type 2 diabetic patients to determine the relationship between serum uric acid (SUA) and NAFLD in type 2 diabetic patients. Clinical parameters including SUA were determined and NAFLD was diagnosed by ultrasonography. SUA was significantly higher in type 2 diabetic subjects with NAFLD than in those without NAFLD in men, but not in women. Furthermore, the prevalence rate of NAFLD increased progressively across the sex-specific SUA tertiles only in men (37.9%, 58.6%, and 72.6%, resp., P for trend < 0.001). After adjusting for confounding factors, the odd ratios (95% CI) for NAFLD were 1 (reference), 2.93 (95%CI 1.25-6.88), and 3.93 (95% CI 1.55-9.98), respectively, across the tertiles of SUA in men. Contrastingly, SUA levels in women were not independently associated with the risk of NAFLD. Our data suggests that SUA is specifically associated with NAFLD in male type 2 diabetic subjects, independent of insulin resistance and other metabolic factors.
Collapse
Affiliation(s)
- Nengguang Fan
- Department of Endocrinology and Metabolism, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Lijuan Zhang
- Department of Endocrinology, Shanghai Songjiang Center Hospital, Shanghai 201600, China
| | - Zhenhua Xia
- Department of Endocrinology, Shanghai Songjiang Center Hospital, Shanghai 201600, China
| | - Liang Peng
- Department of Laboratory Medicine, Shanghai Songjiang Center Hospital, Shanghai 201600, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
- *Yongde Peng:
| |
Collapse
|
456
|
Zhou Y, Wei F, Fan Y. High serum uric acid and risk of nonalcoholic fatty liver disease: A systematic review and meta-analysis. Clin Biochem 2015; 49:636-42. [PMID: 26738417 DOI: 10.1016/j.clinbiochem.2015.12.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/25/2015] [Accepted: 12/26/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Emerging evidence connects serum uric acid (SUA) levels to nonalcoholic fatty liver disease (NAFLD). The objective of this study was to systematically evaluate the association between SUA levels and risk of NAFLD by conducting a meta-analysis of available observational studies. DESIGN AND METHODS We searched for relevant studies in PubMed, Embase, China National Knowledge Infrastructure, and Wanfang databases until October 2014. All observational studies that evaluated SUA levels and NAFLD risks were included. Pooled adjusted odds ratio (OR) and corresponding 95% confidence intervals (CI) were calculated comparing the highest to lowest SUA category. RESULTS Four cross-sectional studies, two prospective studies, and three retrospective studies involving 55,573 participants were identified. In overall risk estimates, the pooled OR of NAFLD occurrence was 1.92 (95% CI: 1.59-2.31) comparing the highest to lowest SUA levels in a random effect model. Subgroup analysis showed that high SUA levels increased the risk of NAFLD in cross-sectional studies (OR: 2.18; 95% CI: 1.58-3.03), retrospective studies (OR 1.82; 95% CI: 1.43-2.33), and prospective studies (OR 1.43; 95% CI: 1.20-1.71). The risk of NAFLD seemed more pronounced among women (OR 1.85; 95% CI: 1.43-2.38) than among men (OR 1.56; 95% CI: 1.30-1.86). CONCLUSION This meta-analysis suggests that increased SUA level is associated with an exacerbated risk of NAFLD. This increased risk is probably independent of conventional NAFLD risk factors.
Collapse
Affiliation(s)
- Yongjing Zhou
- Institute of Molecular Biology & Translational Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, PR China (212002)
| | - Feifei Wei
- Institute of Molecular Biology & Translational Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, PR China (212002)
| | - Yu Fan
- Institute of Molecular Biology & Translational Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, PR China (212002).
| |
Collapse
|
457
|
Xu C. Hyperuricemia and nonalcoholic fatty liver disease: from bedside to bench and back. Hepatol Int 2015; 10:286-93. [DOI: 10.1007/s12072-015-9682-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/22/2015] [Indexed: 12/26/2022]
|
458
|
Testa A, Prudente S, Leonardis D, Spoto B, Sanguedolce MC, Parlongo RM, Tripepi G, Rizza S, Mallamaci F, Federici M, Trischitta V, Zoccali C. A genetic marker of hyperuricemia predicts cardiovascular events in a meta-analysis of three cohort studies in high risk patients. Nutr Metab Cardiovasc Dis 2015; 25:1087-1094. [PMID: 26607700 DOI: 10.1016/j.numecd.2015.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/24/2015] [Accepted: 08/12/2015] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The strongest genetic marker of uric acid levels, the rs734553 SNP in the GLUT9 urate transporter gene, predicts progression to kidney failure in CKD patients and associates with systolic BP and carotid intima media thickness in family-based studies. METHODS Since genes are transmitted randomly (Mendelian randomization) we used this gene polymorphism as an unconfounded research instrument to further explore the link between uric acid and cardiovascular disease (cardiovascular death, and non-fatal myocardial infarction and stroke) in a meta-analysis of three cohort studies formed by high risk patients (MAURO: 755 CKD patients; GHS: 353 type 2 diabetics and coronary artery disease and the TVAS: 119 patients with myocardial infarction). RESULTS In separate analyses of the three cohorts, the incidence rate of CV events was higher in patients with the rs734553 risk (T) allele (TT/GT) than in those without (GG patients) and the HR in TT/GT patients in the three cohorts (range 1.72-2.14) coherently signaled an excessive cardiovascular risk with no heterogeneity (I2 = 0.01). The meta-analytical estimate (total number of patients, n = 1227; total CV events, n = 222) of the HR for the combined end-point in TT/GT patients was twice higher (pooled HR: 2.04, 95% CI: 1.11-3.75, P = 0.02) than in GG homozygotes. CONCLUSIONS The T allele of the rs734553 polymorphism in the GLUT9 gene predicts a doubling in the risk for incident cardiovascular events in patients at high cardiovascular risk. Findings in this study are compatible with the hypothesis of a causal role of hyperuricemia in cardiovascular disease in high risk conditions.
Collapse
Affiliation(s)
- A Testa
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - S Prudente
- IRCCS Casa Sollievo della Sofferenza Mendel Laboratory, San Giovanni Rotondo, Italy
| | - D Leonardis
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - B Spoto
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - M C Sanguedolce
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - R M Parlongo
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - G Tripepi
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - S Rizza
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - F Mallamaci
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy
| | - M Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - V Trischitta
- IRCCS Casa Sollievo della Sofferenza Mendel Laboratory, San Giovanni Rotondo, Italy; Research Unit of Diabetes and Endocrine Diseases IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - C Zoccali
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension, Reggio Calabria, Italy.
| |
Collapse
|
459
|
Dornas WC, de Lima WG, Pedrosa ML, Silva ME. Health implications of high-fructose intake and current research. Adv Nutr 2015; 6:729-37. [PMID: 26567197 PMCID: PMC4642413 DOI: 10.3945/an.114.008144] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although fructose consumption has dramatically increased and is suspected to be causally linked to metabolic abnormalities, the mechanisms involved are still only partially understood. We discuss the available data and investigate the effects of dietary fructose on risk factors associated with metabolic disorders. The evidence suggests that fructose may be a predisposing cause in the development of insulin resistance in association with the induction of hypertriglyceridemia. Experiments in animals have shown this relation when they are fed diets very high in fructose or sucrose, and human studies also show this relation, although with conflicting results due to the heterogeneity of the studies. The link between increased fructose consumption and increases in uric acid also has been confirmed as a potential risk factor for metabolic syndrome, and insulin resistance/hyperinsulinemia may be causally related to the development of hypertension. Collectively, these results suggest a link between high fructose intake and insulin resistance, although future studies must be of reasonable duration, use defined populations, and improve comparisons regarding the effects of relevant doses of nutrients on specific endpoints to fully understand the effect of fructose intake in the absence of potential confounding factors.
Collapse
Affiliation(s)
- Waleska C Dornas
- Research in Biological Sciences-Center for Research in Biological Sciences (NUPEB),
| | - Wanderson G de Lima
- Research in Biological Sciences–Center for Research in Biological Sciences (NUPEB),,Department of Biological Sciences, Institute of Exact and Biological Sciences, and
| | - Maria L Pedrosa
- Research in Biological Sciences–Center for Research in Biological Sciences (NUPEB),,Department of Biological Sciences, Institute of Exact and Biological Sciences, and
| | - Marcelo E Silva
- Research in Biological Sciences–Center for Research in Biological Sciences (NUPEB),,Department of Foods, School of Nutrition, Federal University of Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
460
|
Zelber-Sagi S, Ben-Assuli O, Rabinowich L, Goldstein A, Magid A, Shalev V, Shibolet O, Chodick G. The association between the serum levels of uric acid and alanine aminotransferase in a population-based cohort. Liver Int 2015; 35:2408-15. [PMID: 25845417 DOI: 10.1111/liv.12842] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/29/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Elevated serum uric acid levels reflect and also cause both oxidative stress and insulin resistance and are frequently observed in patients with the metabolic syndrome. A strong association exists between the metabolic syndrome and non-alcoholic fatty liver disease (NAFLD). Therefore, we aimed to test the association between uric acid and elevated alanine aminotransferase (ALT), as a surrogate for NAFLD, using real-world data. METHODS Data used for the cross-sectional study were obtained from Maccabi Healthcare System, a 2-million member health maintenance organization in Israel. The population consisted of individuals aged 20-60 years who underwent blood tests for ALT and uric acid between 1997 and 2012. Individuals with secondary liver disease, celiac, and inflammatory bowel-disease were excluded. Subgroup analysis was performed in subjects who were given the diagnosis of fatty liver in their medical records (n = 2628). RESULTS The study population included 82,608 people (32.5% men, mean age 43.91 ± 10.15 years). There was a significant positive dose-response association between serum uric acid levels and the rate of elevated serum ALT (P for trend <0.001). In multivariable model, controlling for potential confounders, the association between uric acid and elevated ALT persisted (OR = 2.10, 95% CI 1.93-2.29, for the fourth quartile vs. the first). This association was maintained in all categories of gender and BMI. Similar results were observed among patients diagnosed with fatty liver (OR = 1.77, 1.22-2.57). CONCLUSIONS Serum uric acid is independently associated with elevated ALT, as a surrogate for NAFLD, and thus may serve as a serum marker for liver damage and should be further investigated as a risk factor for NAFLD.
Collapse
Affiliation(s)
- Shira Zelber-Sagi
- School of Public Health, University of Haifa, Haifa, Israel.,The Liver Unit, Gastroenterology Department, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - Ofir Ben-Assuli
- Faculty of Business Administration, Ono Academic College, Kiryat Ono, Israel
| | - Liane Rabinowich
- The Liver Unit, Gastroenterology Department, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - Alex Goldstein
- Medical Division, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Avi Magid
- School of Public Health, University of Haifa, Haifa, Israel
| | - Varda Shalev
- Medical Division, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Oren Shibolet
- The Liver Unit, Gastroenterology Department, Tel-Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriel Chodick
- Medical Division, Maccabi Healthcare Services, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
461
|
Di Bartolomeo F, Van den Ende W. Fructose and Fructans: Opposite Effects on Health? PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2015; 70:227-37. [PMID: 25904233 DOI: 10.1007/s11130-015-0485-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Fructans are fructose-based oligo-and polysaccharides of natural origin. Fructan and fructose species are sometimes confused by the great public, although they clearly have different biochemical and physiological properties. This review discusses aspects of the use of fructose and fructans in foods in the context of human health, with possible differential effects on cellular autophagy in cells of the human body. Although there are uncertainties on the daily levels of ingested fructose to be considered harmful to human health, there is an emerging consensus on the benefits of the use of fructans in functional foods, sustaining health via direct immunomodulatory and antioxidant effects or through indirect, prebiotic mechanisms.
Collapse
Affiliation(s)
- Francesca Di Bartolomeo
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/2, 8010, Graz, Austria
| | | |
Collapse
|
462
|
Bjornstad P, Lanaspa MA, Ishimoto T, Kosugi T, Kume S, Jalal D, Maahs DM, Snell-Bergeon JK, Johnson RJ, Nakagawa T. Fructose and uric acid in diabetic nephropathy. Diabetologia 2015; 58:1993-2002. [PMID: 26049401 PMCID: PMC4826347 DOI: 10.1007/s00125-015-3650-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/23/2015] [Indexed: 12/16/2022]
Abstract
Clinical studies have reported associations between serum uric acid levels and the development of diabetic nephropathy, but the underlying mechanisms remain elusive. There is evidence from animal studies that blocking uric acid production protects the kidney from tubulointerstitial injury, which may suggest a causal role for uric acid in the development of diabetic tubular injury. In turn, when fructose, which is endogenously produced in diabetes via the polyol pathway, is metabolised, uric acid is generated from a side-chain reaction driven by ATP depletion and purine nucleotide turnover. For this reason, uric acid derived from endogenous fructose could cause tubulointerstitial injury in diabetes. Accordingly, our research group recently demonstrated that blocking fructose metabolism in a diabetic mouse model mitigated the development of tubulointerstitial injury by lowering tubular uric acid production. In this review we discuss the relationship between uric acid and fructose as a novel mechanism for the development of diabetic tubular injury.
Collapse
Affiliation(s)
- Petter Bjornstad
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO, US
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Takuji Ishimoto
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Diana Jalal
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - David M. Maahs
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO, US
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | | | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, USA
| | - Takahiko Nakagawa
- TMK Project, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
463
|
Tappy L, Lê KA. Health effects of fructose and fructose-containing caloric sweeteners: where do we stand 10 years after the initial whistle blowings? Curr Diab Rep 2015; 15:54. [PMID: 26104800 PMCID: PMC4477723 DOI: 10.1007/s11892-015-0627-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Suspicion that fructose-containing caloric sweeteners (FCCS) may play a causal role in the development of metabolic diseases has elicited intense basic and clinical research over the past 10 years. Prospective cohort studies converge to indicate that FCCS, and more specifically sugar-sweetened beverages (SSBs), consumption is associated with weight gain over time. Intervention studies in which FCCS or SSB consumption is altered while food intake is otherwise left ad libitum indicate that increased FCCS generally increases total energy intake and body weight, while FCCS reduction decreases body weight gain. Clinical trials assessing the effects of SSB reduction as a sole intervention however fail to observe clinically significant weight loss. Many mechanistic studies indicate that excess FCCS can cause potential adverse metabolic effects. Whether this is associated with a long-term risk remains unknown. Scientific evidence that excess FCCS intake causes more deleterious effects to health than excess of other macronutrients is presently lacking. However, the large consumption of FCCS in the population makes it one out of several targets for the treatment and prevention of metabolic diseases.
Collapse
Affiliation(s)
- Luc Tappy
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland,
| | | |
Collapse
|
464
|
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, CO
| | - Tony Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, CO
| |
Collapse
|
465
|
Bortolasci CC, Vargas HO, Vargas Nunes SO, de Melo LGP, de Castro MRP, Moreira EG, Dodd S, Barbosa DS, Berk M, Maes M. Factors influencing insulin resistance in relation to atherogenicity in mood disorders, the metabolic syndrome and tobacco use disorder. J Affect Disord 2015; 179:148-55. [PMID: 25863911 DOI: 10.1016/j.jad.2015.03.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/22/2015] [Accepted: 03/24/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE This study examines the effects of malondialdehyde (MDA) and uric acid on insulin resistance and atherogenicity in subjects with and without mood disorders, the metabolic syndrome (MetS) and tobacco use disorder (TUD). METHODS We included 314 subjects with depression and bipolar depression, with and without the MetS and TUD and computed insulin resistance using the updated homeostasis model assessment (HOMA2IR) and atherogenicity using the atherogenic index of plasma (AIP), that is log10 (triglycerides/high density lipoprotein (HDL) cholesterol. RESULTS HOMA2IR is correlated with body mass index (BMI) and uric acid levels, but not with mood disorders and TUD, while the AIP is positively associated with BMI, mood disorders, TUD, uric acid, MDA and male sex. Uric acid is positively associated with insulin and triglycerides and negatively with HDL cholesterol. MDA is positively associated with triglyceride levels. Comorbid mood disorders and TUD further increase AIP but not insulin resistance. Glucose is positively associated with increasing age, male gender and BMI. DISCUSSION The results show that mood disorders, TUD and BMI together with elevated levels of uric acid and MDA independently contribute to increased atherogenic potential, while BMI and uric acid are risk factors for insulin resistance. The findings show that mood disorders and TUD are closely related to an increased atherogenic potential but not to insulin resistance or the MetS. Increased uric acid is a highly significant risk factor for insulin resistance and increased atherogenic potential. MDA, a marker of lipid peroxidation, further contributes to different aspects of the atherogenic potential. Mood disorders and TUD increase triglyceride levels, lower HDL cholesterol and are strongly associated with the atherogenic, but not insulin resistance, component of the MetS.
Collapse
Affiliation(s)
- Chiara Cristina Bortolasci
- Health Sciences Postgraduate Program, State University of Londrina, Londrina, Paraná, Brazil; Impact Strategic Research Centre, Deakin University, Barwon Health, Geelong, VIC, Australia
| | | | | | - Luiz Gustavo Piccoli de Melo
- Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil
| | - Márcia Regina Pizzo de Castro
- Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil
| | | | - Seetal Dodd
- Impact Strategic Research Centre, Deakin University, Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia
| | - Décio Sabbatini Barbosa
- Health Sciences Postgraduate Program, State University of Londrina, Londrina, Paraná, Brazil; Department of Pathology, Clinical Analysis, and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Michael Berk
- Impact Strategic Research Centre, Deakin University, Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia; Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
| | - Michael Maes
- Health Sciences Postgraduate Program, State University of Londrina, Londrina, Paraná, Brazil; Impact Strategic Research Centre, Deakin University, Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
466
|
Lima WG, Martins-Santos MES, Chaves VE. Uric acid as a modulator of glucose and lipid metabolism. Biochimie 2015; 116:17-23. [PMID: 26133655 DOI: 10.1016/j.biochi.2015.06.025] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/26/2015] [Indexed: 01/05/2023]
Abstract
In humans, uric acid is the final oxidation product of purine catabolism. The serum uric acid level is based on the balance between the absorption, production and excretion of purine. Uric acid is similarly produced in the liver, adipose tissue and muscle and is primarily excreted through the urinary tract. Several factors, including a high-fructose diet and the use of xenobiotics and alcohol, contribute to hyperuricaemia. Hyperuricaemia belongs to a cluster of metabolic and haemodynamic abnormalities, called metabolic syndrome, characterised by abdominal obesity, glucose intolerance, insulin resistance, dyslipidaemia and hypertension. Hyperuricaemia reduction in the Pound mouse or fructose-fed rats, as well as hyperuricaemia induction by uricase inhibition in rodents and studies using cell culture have suggested that uric acid plays an important role in the development of metabolic syndrome. These studies have shown that high uric acid levels regulate the oxidative stress, inflammation and enzymes associated with glucose and lipid metabolism, suggesting a mechanism for the impairment of metabolic homeostasis. Humans lacking uricase, the enzyme responsible for uric acid degradation, are susceptible to these effects. In this review, we summarise the current knowledge of the effects of uric acid on the regulation of metabolism, primarily focusing on liver, adipose tissue and skeletal muscle.
Collapse
Affiliation(s)
- William Gustavo Lima
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Valéria Ernestânia Chaves
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil.
| |
Collapse
|
467
|
Aroor AR, Habibi J, Ford DA, Nistala R, Lastra G, Manrique C, Dunham MM, Ford KD, Thyfault JP, Parks EJ, Sowers JR, Rector RS. Dipeptidyl peptidase-4 inhibition ameliorates Western diet-induced hepatic steatosis and insulin resistance through hepatic lipid remodeling and modulation of hepatic mitochondrial function. Diabetes 2015; 64:1988-2001. [PMID: 25605806 PMCID: PMC4439570 DOI: 10.2337/db14-0804] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023]
Abstract
Novel therapies are needed for treating the increasing prevalence of hepatic steatosis in Western populations. In this regard, dipeptidyl peptidase-4 (DPP-4) inhibitors have recently been reported to attenuate the development of hepatic steatosis, but the potential mechanisms remain poorly defined. In the current study, 4-week-old C57Bl/6 mice were fed a high-fat/high-fructose Western diet (WD) or a WD containing the DPP-4 inhibitor, MK0626, for 16 weeks. The DPP-4 inhibitor prevented WD-induced hepatic steatosis and reduced hepatic insulin resistance by enhancing insulin suppression of hepatic glucose output. WD-induced accumulation of hepatic triacylglycerol (TAG) and diacylglycerol (DAG) content was significantly attenuated with DPP-4 inhibitor treatment. In addition, MK0626 significantly reduced mitochondrial incomplete palmitate oxidation and increased indices of pyruvate dehydrogenase activity, TCA cycle flux, and hepatic TAG secretion. Furthermore, DPP-4 inhibition rescued WD-induced decreases in hepatic PGC-1α and CPT-1 mRNA expression and hepatic Sirt1 protein content. Moreover, plasma uric acid levels in mice fed the WD were decreased after MK0626 treatment. These studies suggest that DPP-4 inhibition ameliorates hepatic steatosis and insulin resistance by suppressing hepatic TAG and DAG accumulation through enhanced mitochondrial carbohydrate utilization and hepatic TAG secretion/export with a concomitant reduction of uric acid production.
Collapse
Affiliation(s)
- Annayya R Aroor
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, MO Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO
| | - Javad Habibi
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, MO Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO Center for Cardiovascular Research, Saint Louis University, St. Louis, MO
| | - Ravi Nistala
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, MO Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO
| | - Guido Lastra
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, MO Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO
| | - Camila Manrique
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, MO Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO
| | - Merlow M Dunham
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO Center for Cardiovascular Research, Saint Louis University, St. Louis, MO
| | - Kaitlin D Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO Center for Cardiovascular Research, Saint Louis University, St. Louis, MO
| | - John P Thyfault
- Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - Elizabeth J Parks
- Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - James R Sowers
- Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Missouri, Columbia, MO Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO
| | - R Scott Rector
- Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| |
Collapse
|
468
|
Yracheta JM, Lanaspa MA, Le MT, Abdelmalak MF, Alfonso J, Sánchez-Lozada LG, Johnson RJ. Diabetes and Kidney Disease in American Indians: Potential Role of Sugar-Sweetened Beverages. Mayo Clin Proc 2015; 90:813-23. [PMID: 26046414 DOI: 10.1016/j.mayocp.2015.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/19/2015] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
Since the early 20th century, a marked increase in obesity, diabetes, and chronic kidney disease has occurred in the American Indian population, especially the Pima Indians of the Southwest. Here, we review the current epidemic and attempt to identify remediable causes. A search was performed using PubMed and the search terms American Indian and obesity, American Indian and diabetes, American Indian and chronic kidney disease, and American Indian and sugar or fructose, Native American, Alaska Native, First Nations, Aboriginal, Amerind, and Amerindian for American Indian for articles linking American Indians with diabetes, obesity, chronic kidney disease, and sugar; additional references were identified in these publications traced to 1900 and articles were reviewed if they were directly discussing these topics. Multiple factors are involved in the increased risk for diabetes and kidney disease in the American Indian population, including poverty, overnutrition, poor health care, high intake of sugar, and genetic mechanisms. Genetic factors may be especially important in the Pima, as historical records suggest that this group was predisposed to obesity before exposure to Western culture and diet. Exposure to sugar-sweetened beverages may also be involved in the increased risk for chronic kidney disease. In these small populations in severe health crisis, we recommend further studies to investigate the role of excess added sugar, especially sugar-sweetened beverages, as a potentially remediable risk factor.
Collapse
Affiliation(s)
- Joseph M Yracheta
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA.
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, CO; Colorado Research Partners LLC, Aurora, CO
| | - MyPhuong T Le
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, CO; Colorado Research Partners LLC, Aurora, CO
| | - Manal F Abdelmalak
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC
| | - Javier Alfonso
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, CO
| | - Laura G Sánchez-Lozada
- Colorado Research Partners LLC, Aurora, CO; Laboratory of Renal Physiopathology and Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado, Aurora, CO; Colorado Research Partners LLC, Aurora, CO
| |
Collapse
|
469
|
Nutrigenomics analysis reveals that copper deficiency and dietary sucrose up-regulate inflammation, fibrosis and lipogenic pathways in a mature rat model of nonalcoholic fatty liver disease. J Nutr Biochem 2015; 26:996-1006. [PMID: 26033743 DOI: 10.1016/j.jnutbio.2015.04.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 04/08/2015] [Accepted: 04/13/2015] [Indexed: 12/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) prevalence is increasing worldwide, with the affected US population estimated near 30%. Diet is a recognized risk factor in the NAFLD spectrum, which includes nonalcoholic steatohepatitis (NASH) and fibrosis. Low hepatic copper (Cu) was recently linked to clinical NAFLD/NASH severity. Simple sugar consumption including sucrose and fructose is implicated in NAFLD, while consumption of these macronutrients also decreases liver Cu levels. Though dietary sugar and low Cu are implicated in NAFLD, transcript-level responses that connect diet and pathology are not established. We have developed a mature rat model of NAFLD induced by dietary Cu deficiency, human-relevant high sucrose intake (30% w/w) or both factors in combination. Compared to the control diet with adequate Cu and 10% (w/w) sucrose, rats fed either high-sucrose or low-Cu diet had increased hepatic expression of genes involved in inflammation and fibrogenesis, including hepatic stellate cell activation, while the combination of diet factors also increased ATP citrate lyase and fatty acid synthase gene transcription (fold change > 2, P < 0.02). Low dietary Cu decreased hepatic and serum Cu (P ≤ 0.05), promoted lipid peroxidation and induced NAFLD-like histopathology, while the combined factors also induced fasting hepatic insulin resistance and liver damage. Neither low Cu nor 30% sucrose in the diet led to enhanced weight gain. Taken together, transcript profiles, histological and biochemical data indicate that low Cu and high sucrose promote hepatic gene expression and physiological responses associated with NAFLD and NASH, even in the absence of obesity or severe steatosis.
Collapse
|
470
|
Donderski R, Miśkowiec-Wiśniewska I, Kretowicz M, Grajewska M, Manitius J, Kamińska A, Junik R, Siódmiak J, Stefańska A, Odrowąż-Sypniewska G, Pluta A, Lanaspa M, Johnson RJ. The fructose tolerance test in patients with chronic kidney disease and metabolic syndrome in comparison to healthy controls. BMC Nephrol 2015; 16:68. [PMID: 25935771 PMCID: PMC4450852 DOI: 10.1186/s12882-015-0048-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 04/01/2015] [Indexed: 01/08/2023] Open
Abstract
Background Fructose acutely raises serum uric acid in normal subjects, but the effect in subjects with metabolic syndrome or subjects with chronic kidney disease is unknown. The aim of the study was to evaluate changes in serum uric acid during the fructose tolerance test in patients with chronic kidney disease, metabolic syndrome with comparison to healthy controls. Methods Studies were performed in 36 subjects with obesity (body mass index >30) and metabolic syndrome, 14 patients with stage 3 chronic kidney disease, and 25 healthy volunteers. The fructose tolerance test was performed in each patient. The change in serum uric acid during the fructose challenge was correlated with baseline ambulatory blood pressure, serum uric acid, metabolic, and inflammatory markers, and target organ injury including carotid intima media thickness and renal resistive index (determined by Doppler). Results Absolute serum uric acid values were highest in the chronic kidney disease group, followed by the metabolic syndrome and then healthy controls. Similar increases in serum uric acid in response to the fructose tolerance test was observed in all three groups, but the greatest percent rise was observed in healthy controls compared to the other two groups. No significant association was shown between the relative rise in uric acid and clinical or inflammatory parameters associated with kidney disease (albuminuria, eGFR) or metabolic syndrome. Conclusions Subjects with chronic kidney disease and metabolic syndrome have higher absolute uric acid values following a fructose tolerance test, but show a relatively smaller percent increase in serum uric acid. Changes in serum uric acid during the fructose tolerance test did not correlate with changes in metabolic parameters, inflammatory mediators or with target organ injury. These studies suggest that acute changes in serum uric acid in response to fructose do not predict the metabolic phenotype or presence of inflammatory mediators in subjects with obesity, metabolic syndrome or chronic kidney disease. Trial registration The study was registered in ClinicalTrials.gov. Identifier : NCT01332526. www.register.clinicaltrials.gov/01332526 Electronic supplementary material The online version of this article (doi:10.1186/s12882-015-0048-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rafał Donderski
- Department of Nephrology, Hypertension and Internal Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Ilona Miśkowiec-Wiśniewska
- Department of Nephrology, Hypertension and Internal Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Marek Kretowicz
- Department of Nephrology, Hypertension and Internal Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Magdalena Grajewska
- Department of Nephrology, Hypertension and Internal Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Jacek Manitius
- Department of Nephrology, Hypertension and Internal Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Anna Kamińska
- Department of Diabetology and Endocrinology, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Roman Junik
- Department of Diabetology and Endocrinology, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Joanna Siódmiak
- Department of Laboratory Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Anna Stefańska
- Department of Laboratory Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Grażyna Odrowąż-Sypniewska
- Department of Laboratory Medicine, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Agnieszka Pluta
- Institute of Public Nursing, Collegium Medicum, Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland.
| | - Miguel Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Denver, CO, USA.
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Denver, CO, USA.
| |
Collapse
|
471
|
Wu SJ, Zhu GQ, Ye BZ, Kong FQ, Zheng ZX, Zou H, Shi KQ, Lin L, Braddock M, Huang WJ, Chen YP, Zheng MH. Association between sex-specific serum uric acid and non-alcoholic fatty liver disease in Chinese adults: a large population-based study. Medicine (Baltimore) 2015; 94:e802. [PMID: 25929934 PMCID: PMC4603030 DOI: 10.1097/md.0000000000000802] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The aim of this study was to examine the association between sex-specific serum uric acid (sUA) levels and NAFLD in a large population-based study.A total of 60,455 subjects from 2 separate medical centers were included. Sex-specific sUA quartiles (Q1-Q4) were defined: ≤330, 331-380, 381-435, and ≥436 μmol/L for male; ≤230, 231-270, 271-310, and ≥311 μmol/L for female. The odds ratios (ORs), hazard ratios (HRs), and 95% confidence intervals (CIs) for NAFLD were calculated across each quartile of sUA, using the Q1 as reference.After adjusting for known confounding variables in this study, the ORs for NAFLD in the cross-sectional population were 1.211 (95% CI 1.109-1.322), 1.519 (95% CI 1.395-1.654), 1.903 (95% CI 1.748-2.072) for Q2, Q3, and Q4, respectively. In the longitudinal population, compared with the reference group, those in Q2, Q3, and Q4 had HRs of 1.127 (95% CI 0.956-1.330), 1.380 (95% CI 1.157-1.644), 1.589 (95% CI 1.310-1.927) for NAFLD, respectively. Analysis for the sex-specific subgroup showed the adjusted ORs for Q4 versus Q1 were 2.898 (95% CI 2.36-3.588) in female and 1.887 (95% CI 1.718-2.072) in male in the cross-sectional population. In the longitudinal population, the HRs for the Q4 were 2.355 (95% CI 1.702-3.259) in female and 1.249 (95% CI 0.975-1.601) in male, compared with Q1.We report that a sex-specific sUA level is independently associated with NAFLD. The association between sUA and NAFLD was significantly greater in females than in males.
Collapse
Affiliation(s)
- Sheng-Jie Wu
- From the Department of Cardiovascular Medicine, the Heart Center (S-JW, B-ZY, F-QK, Z-XZ, LL, W-JH); Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University (G-QZ, K-QS, Y-PC, M-HZ); School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou (G-QZ); Department of Internal Medicine, Xinyu People's Hospital of Jiangxi Province, Xinyu (HZ); Institute of Hepatology, Wenzhou Medical University, Wenzhou, China (K-QS, Y-PC, M-HZ); and Global Medicines Development, AstraZeneca R&D, Loughborough, United Kingdom (MB)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
472
|
Himes KP, Young A, Koppes E, Stolz D, Barak Y, Sadovsky Y, Chaillet JR. Loss of inherited genomic imprints in mice leads to severe disruption in placental lipid metabolism. Placenta 2015; 36:389-96. [PMID: 25662615 PMCID: PMC4359963 DOI: 10.1016/j.placenta.2015.01.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/29/2014] [Accepted: 01/22/2015] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Monoallelic expression of imprinted genes is necessary for placental development and normal fetal growth. Differentially methylated domains (DMDs) largely determine the parental-specific monoallelic expression of imprinted genes. Maternally derived DNA (cytosine-5-) -methyltransferase 1o (DNMT1o) maintains DMDs during the eight-cell stage of development. DNMT1o-deficient mouse placentas have a generalized disruption of genomic imprints. Previous studies have demonstrated that DNMT1o deficiency alters placental morphology and broadens the embryonic weight distribution in late gestation. Lipids are critical for fetal growth. Thus, we assessed the impact of disrupted imprinting on placental lipids. METHODS Lipids were quantified from DNMT1o-deficient mouse placentas and embryos at E17.5 using a modified Folch method. Expression of select genes critical for lipid metabolism was quantified with RT-qPCR. Mitochondrial morphology was assessed by TEM and mitochondrial aconitase and cytoplasmic citrate concentrations quantified. DMD methylation was determined by EpiTYPER. RESULTS We found that DNMT1o deficiency is associated with increased placental triacylglycerol levels. Neither fetal triacylglycerol concentrations nor expression of select genes that mediate placental lipid transport were different from wild type. Placental triacylglycerol accumulation was associated with impaired beta-oxidation and abnormal citrate metabolism with decreased mitochondrial aconitase activity and increased cytoplasmic citrate concentrations. Loss of methylation at the MEST DMD was strongly associated with placental triacylglycerol accumulation. DISCUSSION A generalized disruption of genomic imprints leads to triacylglycerol accumulation and abnormal mitochondrial function. This could stem directly from a loss of methylation at a given DMD, such as MEST, or represent a consequence of abnormal placental development.
Collapse
Affiliation(s)
- K P Himes
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| | - A Young
- Magee-Womens Research Institute, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| | - E Koppes
- Magee-Womens Research Institute, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| | - D Stolz
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Y Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA; Magee-Womens Research Institute, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| | - Y Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA; Magee-Womens Research Institute, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| | - J R Chaillet
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, 204 Craft Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
473
|
Yracheta JM, Alfonso J, Lanaspa MA, Roncal-Jimenez C, Johnson SB, Sánchez-Lozada LG, Johnson RJ. Hispanic Americans living in the United States and their risk for obesity, diabetes and kidney disease: Genetic and environmental considerations. Postgrad Med 2015; 127:503-10. [PMID: 25746679 DOI: 10.1080/00325481.2015.1021234] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Hispanic American, the largest minority population in the United States, is at increased risk for obesity, diabetes and end-stage renal disease. Here we review genetic and environmental factors that might account for their increased risk for these conditions. Whereas many environmental and genetic factors have important roles in driving the increased risk for obesity and kidney disease in this population, a case is made that excessive intake of sugary beverages is a contributory cause. Studies focusing on decreasing intake of sugary beverages among the Hispanic American could potentially reduce renal and cardiovascular complications in this population.
Collapse
Affiliation(s)
- Joseph M Yracheta
- Department of Pharmaceutics, School of Pharmacy, University of Washington , Seattle, WA , USA
| | | | | | | | | | | | | |
Collapse
|
474
|
Interleukin-32γ attenuates ethanol-induced liver injury by the inhibition of cytochrome P450 2E1 expression and inflammatory responses. Clin Sci (Lond) 2015; 128:695-706. [DOI: 10.1042/cs20140576] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol abuse and alcoholism lead to alcoholic liver disease (ALD), which is a major type of chronic liver disease worldwide. Interleukin-32 (IL-32) is a novel cytokine involved in inflammation and cancer development. However, the role of IL-32 in chronic liver disease has not been reported. In the present paper, we tested the effect of IL-32γ on ethanol-induced liver injury in IL-32γ-overexpressing transgenic mice (IL-32γ mice) after chronic ethanol feeding. Male C57BL/6 and IL-32γ mice (10–12 weeks old) were fed on a Lieber–DeCarli diet containing 6.6% ethanol for 6 weeks. IL-32γ-transfected HepG2 and Huh7 cells, as well as primary hepatocytes from IL-32γ mice, were treated with or without ethanol. The hepatic steatosis and damage induced by ethanol administration were attenuated in IL-32γ mice. Ethanol-induced cytochrome P450 2E1 expression and hydrogen peroxide levels were decreased in the livers of IL-32γ mice, primary hepatocytes from IL-32γ mice and IL-32γ-overexpressing human hepatic cells. The ethanol-induced expression levels of cyclo-oxygenase-2 (COX-2) and IL-6 were reduced in the livers of IL-32γ mice. Because nuclear transcription factor κB (NF-κB) is a key redox transcription factor of inflammatory responses, we examined NF-κB activity. Ethanol-induced NF-κB activities were significantly lower in the livers of IL-32γ mice than in wild-type (WT) mice. Furthermore, reduced infiltration of natural killer cells, cytotoxic T-cells and macrophages in the liver after ethanol administration was observed in IL-32γ mice. These data suggest that IL-32γ prevents ethanol-induced hepatic injury via the inhibition of oxidative damage and inflammatory responses.
Collapse
|
475
|
Saab KR, Kendrick J, Yracheta JM, Lanaspa MA, Pollard M, Johnson RJ. New insights on the risk for cardiovascular disease in African Americans: the role of added sugars. J Am Soc Nephrol 2015; 26:247-57. [PMID: 25090991 PMCID: PMC4310665 DOI: 10.1681/asn.2014040393] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/30/2014] [Indexed: 12/26/2022] Open
Abstract
African Americans are at increased risk for cardiovascular and metabolic diseases, including obesity, high BP, diabetes, CKD, myocardial infarction, and stroke. Here we summarize the current risks and provide an overview of the underlying risk factors that may account for these associations. By reviewing the relationship between cardiovascular and renal diseases and the African-American population during the early 20th century, the historic and recent associations of African heritage with cardiovascular disease, and modern population genetics, it is possible to assemble strong hypotheses for the primary underlying mechanisms driving the increased frequency of disease in African Americans. Our studies suggest that underlying genetic mechanisms may be responsible for the increased frequency of high BP and kidney disease in African Americans, with particular emphasis on the role of APOL1 polymorphisms in causing kidney disease. In contrast, the Western diet, particularly the relatively high intake of fructose-containing sugars and sweetened beverages, appears to be the dominant force driving the increased risk of diabetes, obesity, and downstream complications. Given that intake of added sugars is a remediable risk factor, we recommend clinical trials to examine the reduction of sweetened beverages as a primary means for reducing cardiovascular risk in African Americans.
Collapse
Affiliation(s)
- Karim R Saab
- Renal Division, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Jessica Kendrick
- Renal Division, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Joseph M Yracheta
- Department of Pharmaceutics, University of Washington, School of Pharmacy, Seattle, Washington
| | - Miguel A Lanaspa
- Renal Division, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado; Colorado Research Partners LLC, Aurora, Colorado; and
| | | | - Richard J Johnson
- Renal Division, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado; Colorado Research Partners LLC, Aurora, Colorado; and
| |
Collapse
|
476
|
Liu P, Wang H, Zhang F, Chen Y, Wang D, Wang Y. The Effects of Allopurinol on the Carotid Intima-media Thickness in Patients with Type 2 Diabetes and Asymptomatic Hyperuricemia: A Three-year Randomized Parallel-controlled Study. Intern Med 2015; 54:2129-37. [PMID: 26328636 DOI: 10.2169/internalmedicine.54.4310] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate the long-term effective control of serum uric acid by allopurinol on the carotid intima-media thickness (IMT) in patients with type 2 diabetes (T2DM) and asymptomatic hyperuricemia (HUA). METHODS This was a randomized open parallel-controlled study. In this study, 176 patients with T2DM and asymptomatic HUA were randomly allocated to the conventional or allopurinol treatment groups on the basis of a computer-generated random number table. Changes in the carotid IMT, biochemical indexes, high sensitive C-reactive protein (hs-CRP) and the incidence of hypertension in patients before and after three years of treatment were examined and compared between the groups. RESULTS There were no statistically significant differences in the baseline characteristics of the study participants between the two treatment groups (p>0.05 for all). Nevertheless, the serum uric acid, triglyceride, and hs-CRP levels and the homeostasis assessment for insulin resistance (HOMA-IR), systolic blood pressure, diastolic blood pressure and the carotid IMT in the allopurinol group were significantly lower than those in the conventional group after three years of treatment (p<0.01 for all). The intention-to-treat analysis indicated that the incidence of new-onset hypertension in the allopurinol group showed a declining trend compared to that in the conventional treatment group (6.8% vs. 13.6%, p>0.05). CONCLUSION The long-term effective control of serum uric acid by allopurinol may improve insulin resistance, decrease the serum levels of hs-CRP, reduce the carotid IMT, and may delay the development of atherosclerosis in patients with T2DM and asymptomatic HUA.
Collapse
Affiliation(s)
- Peng Liu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, China
| | | | | | | | | | | |
Collapse
|
477
|
Abstract
In gouty patients, urate lowering therapies (ULTs) are recommended to bring serum uric acid (SUA) levels below 6.0 mg/dL, with the aim of dissolving urate depositions, thereby reducing disease impact. However, patients with hyperuricemia often present with other conditions associated with cardiovascular (CV) risk, such as high blood pressure, obesity, insulin resistance, fatty liver, and chronic kidney disease. In the last decade, several well grounded pieces of evidence showed that the elevation of uric acid often occurs prior to the development of hypertension or metabolic syndrome, thus suggesting a direct association between elevated SUA and these conditions. This paper will discuss available evidence supporting the key role of serum uric acid in the development of CV and renal disease, with a focus on the molecular mechanisms underlying this causative association. This review is based on a PubMed/Embase database search for articles on hyperuricemia and its impact on cardiovascular and renal function.
Collapse
|
478
|
Moore JB, Gunn PJ, Fielding BA. The role of dietary sugars and de novo lipogenesis in non-alcoholic fatty liver disease. Nutrients 2014; 6:5679-703. [PMID: 25514388 PMCID: PMC4276992 DOI: 10.3390/nu6125679] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023] Open
Abstract
Dietary sugar consumption, in particular sugar-sweetened beverages and the monosaccharide fructose, has been linked to the incidence and severity of non-alcoholic fatty liver disease (NAFLD). Intervention studies in both animals and humans have shown large doses of fructose to be particularly lipogenic. While fructose does stimulate de novo lipogenesis (DNL), stable isotope tracer studies in humans demonstrate quantitatively that the lipogenic effect of fructose is not mediated exclusively by its provision of excess substrates for DNL. The deleterious metabolic effects of high fructose loads appear to be a consequence of altered transcriptional regulatory networks impacting intracellular macronutrient metabolism and altering signaling and inflammatory processes. Uric acid generated by fructose metabolism may also contribute to or exacerbate these effects. Here we review data from human and animal intervention and stable isotope tracer studies relevant to the role of dietary sugars on NAFLD development and progression, in the context of typical sugar consumption patterns and dietary recommendations worldwide. We conclude that the use of hypercaloric, supra-physiological doses in intervention trials has been a major confounding factor and whether or not dietary sugars, including fructose, at typically consumed population levels, effect hepatic lipogenesis and NAFLD pathogenesis in humans independently of excess energy remains unresolved.
Collapse
Affiliation(s)
- J Bernadette Moore
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK.
| | - Pippa J Gunn
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK.
| | - Barbara A Fielding
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK.
| |
Collapse
|
479
|
Stenvinkel P. Obesity--a disease with many aetiologies disguised in the same oversized phenotype: has the overeating theory failed? Nephrol Dial Transplant 2014; 30:1656-64. [PMID: 25361999 DOI: 10.1093/ndt/gfu338] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 09/22/2014] [Indexed: 02/07/2023] Open
Abstract
Evolution has led to metabolic thrift in humans--a genetic heritage that, when exposed to the modern 'obesogenic' milieu with energy-dense food and a sedentary lifestyle, predisposes to obesity. The current paradigm that overeating of easily digestible carbohydrates and the resulting imbalance between energy in and out as the cause of overweight has recently been challenged. Indeed, studies suggest that the host response to various nutrients contributes to overeating and fat accumulation. Alterations in neurotransmitter functions, changes in the epigenome, dysbiosis of gut microbiota and effects of specific nutrients (or lack of such nutrients) on mitochondrial function and signalling pathways may promote fat accumulation independent of calories. Whereas nutrients that stimulate generation of uric acid (such as fructose and purine-rich food) cause insulin resistance and fat accumulation, other nutrients (such as antioxidants, plant food, probiotics, nuts, soy and omega-3) counteract the negative effects of a calorie-rich diet by salutary effects on mitochondrial biogenesis. Thus, the specific metabolic effects of different nutrients may be more important than its total energy content. By studying the impact of nutrients on mitochondrial health, as well as the trans-generational impact of nutrients during fetal life, and how specific bacterial species correlate with fat mass accumulation, new dietary targets for obesity management may emerge. Overeating and overshooting of calories could to a large extent represent a symptom rather than a cause of obesity; therefore, hypocaloric diets should probably not be the main, and certainly not the only, focus for treatment of the obese patient.
Collapse
Affiliation(s)
- Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
480
|
Reduced oxidative stress contributes to the lipid lowering effects of isoquercitrin in free fatty acids induced hepatocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:313602. [PMID: 25404990 PMCID: PMC4227458 DOI: 10.1155/2014/313602] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 07/03/2014] [Indexed: 02/06/2023]
Abstract
Oxidative stress interferes with hepatic lipid metabolism at various levels ranging from benign lipid storage to so-called second hit of inflammation activation. Isoquercitrin (IQ) is widely present flavonoid but its effects on hepatic lipid metabolism remain unknown. We used free fatty acids (FFA) induced lipid overload and oxidative stress model in two types of liver cells and measured cell viability, intracellular lipids, and reactive oxygen species (ROS) within hepatocytes. In addition, Intracellular triglycerides (TG), superoxide dismutase (SOD), and malondialdehyde (MDA) were examined. A novel in vitro model was used to evaluate correlation between lipid lowering and antioxidative activities. Furthermore, 34 major cytokines and corresponding ROS levels were analyzed in FFA/LPS induced coculture model between hepatocytes and Kupffer cells. At molecular level AMPK pathway was elucidated. We showed that IQ attenuated FFA induced lipid overload and ROS within hepatocytes. Further, IQ reversed FFA induced increase in intracellular TG SOD and MDA. It was shown that antioxidative activity of IQ correlates with its lipid lowering potentials. IQ reversed major proinflammatory cytokines and oxidative stress in FFA/LPS induced coculture model. Finally, AMPK pathway was found responsible for metabolic benefits at molecular level. IQ strikingly manifests antioxidative and related lipid lowering activities in hepatocytes.
Collapse
|
481
|
Olea E, Agapito MT, Gallego-Martin T, Rocher A, Gomez-Niño A, Obeso A, Gonzalez C, Yubero S. Intermittent hypoxia and diet-induced obesity: effects on oxidative status, sympathetic tone, plasma glucose and insulin levels, and arterial pressure. J Appl Physiol (1985) 2014; 117:706-19. [DOI: 10.1152/japplphysiol.00454.2014] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Obstructive sleep apnea (OSA) consists of sleep-related repetitive obstructions of upper airways that generate episodes of recurrent or intermittent hypoxia (IH). OSA commonly generates cardiovascular and metabolic pathologies defining the obstructive sleep apnea syndrome (OSAS). Literature usually links OSA-associated pathologies to IH episodes that would cause an oxidative status and a carotid body-mediated sympathetic hyperactivity. Because cardiovascular and metabolic pathologies in obese patients and those with OSAS are analogous, we used models (24-wk-old Wistar rats) of IH (applied from weeks 22 to 24) and diet-induced obesity (O; animals fed a high-fat diet from weeks 12 to 24) to define the effect of each individual maneuver and their combination on the oxidative status and sympathetic tone of animals, and to quantify cardiovascular and metabolic parameters and their deviation from normality. We found that IH and O cause an oxidative status (increased lipid peroxides and diminished activities of superoxide dismutases), an inflammatory status (augmented C-reactive protein and nuclear factor kappa-B activation), and sympathetic hyperactivity (augmented plasma and renal artery catecholamine levels and synthesis rate); combined treatments worsened those alterations. IH and O augmented liver lipid content and plasma cholesterol, triglycerides, leptin, glycemia, insulin levels, and HOMA index, and caused hypertension; most of these parameters were aggravated when IH and O were combined. IH diminished ventilatory response to hypoxia, and hypercapnia and O created a restrictive ventilatory pattern; a combination of treatments led to restrictive hypoventilation. Data demonstrate that IH and O cause comparable metabolic and cardiovascular pathologies via misregulation of the redox status and sympathetic hyperactivity.
Collapse
Affiliation(s)
- Elena Olea
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Maria Teresa Agapito
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Teresa Gallego-Martin
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Asuncion Rocher
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Angela Gomez-Niño
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Ana Obeso
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Constancio Gonzalez
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| | - Sara Yubero
- Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, School of Medicine, Vallodolid, Spain; and Institute of Molecular Biology and Genetics, CIBER Enfermedades Respiratorias, CIBERES, Institute of Salud Carlos III, Vallodolid, Spain
| |
Collapse
|
482
|
Choi YJ, Shin HS, Choi HS, Park JW, Jo I, Oh ES, Lee KY, Lee BH, Johnson RJ, Kang DH. Uric acid induces fat accumulation via generation of endoplasmic reticulum stress and SREBP-1c activation in hepatocytes. J Transl Med 2014; 94:1114-25. [PMID: 25111690 DOI: 10.1038/labinvest.2014.98] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 05/13/2014] [Accepted: 06/10/2014] [Indexed: 12/19/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently one of the most common types of chronic liver injury. Elevated serum uric acid is a strong predictor of the development of fatty liver as well as metabolic syndrome. Here we demonstrate that uric acid induces triglyceride accumulation by SREBP-1c activation via induction of endoplasmic reticulum (ER) stress in hepatocytes. Uric acid-induced ER stress resulted in an increase of glucose-regulated protein (GRP78/94), splicing of the X-box-binding protein-1 (XBP-1), the phosphorylation of protein kinase RNA-like ER kinase (PERK), and eukaryotic translation initiation factor-2α (eIF-2α) in cultured hepatocytes. Uric acid promoted hepatic lipogenesis through overexpression of the lipogenic enzyme, acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS), and stearoyl-CoA desaturase 1 (SCD1) via activation of SREBP-1c, which was blocked by probenecid, an organic anion transport blocker in HepG2 cells and primary hepatocytes. A blocker of ER stress, tauroursodeoxycholic acid (TUDCA), and an inhibitor of SREBP-1c, metformin, blocked hepatic fat accumulation, suggesting that uric acid promoted fat synthesis in hepatocytes via ER stress-induced activation of SREBP-1c. Uric acid-induced activation of NADPH oxidase preceded ER stress, which further induced mitochondrial ROS production in hepatocytes. These studies provide new insights into the mechanisms by which uric acid stimulates fat accumulation in the liver.
Collapse
Affiliation(s)
- Yea-Jin Choi
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| | - Hyun-Soo Shin
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| | - Hack Sun Choi
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| | - Eok-Soo Oh
- Department of Life Sciences, Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Kang-Yo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Richard J Johnson
- The Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado, USA
| | - Duk-Hee Kang
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Republic of Korea
| |
Collapse
|
483
|
Kang DH, Ha SK. Uric Acid Puzzle: Dual Role as Anti-oxidantand Pro-oxidant. Electrolyte Blood Press 2014; 12:1-6. [PMID: 25061467 PMCID: PMC4105384 DOI: 10.5049/ebp.2014.12.1.1] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 06/22/2014] [Indexed: 12/22/2022] Open
Abstract
Hyperuricemia is known to be associated with the presence of cardiovascular and metabolic syndrome and with the development of incipient kidney disease and an accelerated renal progression. However, an elevated uric acid level was not generally regarded as a true etiology or mediator, but an indicator of these diseases. Uric acid has recently regained the clinical interest and popularity based on emerging data suggesting the causative role of hyperuricemia in cardiovascular and renal disease. Experimental data demonstrates oxidative stress is one of the earliest phenomena observed in vascular, renal, liver cells and adipocytes exposed to uric acid. Since uric acid is one of the major antioxidants of plasma acting as a free radical scavenger and a chelator of transitional metal ion, uric acid-induced oxidative stress seems paradoxical. Data regarding the clinical implication of hyperuricemia is even more confusing, which defines hyperuricemia as a useless parameter to be eliminated from routine follow-up or a major risk factor to be therapeutic target. With a review of experimental and epidemiologic data, the presence of molecular switch to regulate the role of uric acid as anti- or pro-oxidant in different compartment of our body is suggested, which may shed light on understanding the paradoxical role of uric acid and solving the "uric acid debate".
Collapse
Affiliation(s)
- Duk-Hee Kang
- Division of Nephrology, Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea. ; Ewha Medical Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Sung-Kyu Ha
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Seoul, Korea
| |
Collapse
|
484
|
Johnson RJ, Nakagawa T, Sánchez-Lozada LG, Lanaspa MA, Tamura Y, Tanabe K, Ishimoto T, Thomas J, Inaba S, Kitagawa W, Rivard CJ. Umami: the taste that drives purine intake. J Rheumatol 2014; 40:1794-6. [PMID: 24187156 DOI: 10.3899/jrheum.130531] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Richard J Johnson
- Division of Kidney Diseases and Hypertension, University of Colorado Denver
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
485
|
Lanaspa MA, Ishimoto T, Cicerchi C, Tamura Y, Roncal-Jimenez CA, Chen W, Tanabe K, Andres-Hernando A, Orlicky DJ, Finol E, Inaba S, Li N, Rivard CJ, Kosugi T, Sanchez-Lozada LG, Petrash JM, Sautin YY, Ejaz AA, Kitagawa W, Garcia GE, Bonthron DT, Asipu A, Diggle CP, Rodriguez-Iturbe B, Nakagawa T, Johnson RJ. Endogenous fructose production and fructokinase activation mediate renal injury in diabetic nephropathy. J Am Soc Nephrol 2014; 25:2526-38. [PMID: 24876114 DOI: 10.1681/asn.2013080901] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Miguel A Lanaspa
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado;
| | - Takuji Ishimoto
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Christina Cicerchi
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Yoshifuru Tamura
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Carlos A Roncal-Jimenez
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Wei Chen
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Katsuyuki Tanabe
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Ana Andres-Hernando
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - David J Orlicky
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Esteban Finol
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado; Venezuelan Scientific Research Institute and University Hospital of Zulia, Maracaibo, Venezuela
| | - Shinichiro Inaba
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Nanxing Li
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Christopher J Rivard
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Laura G Sanchez-Lozada
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado; Laboratory of Renal Physiopathology and Department of Nephrology, INC Ignacio Chavez, Mexico City, Mexico
| | - J Mark Petrash
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | | | - A Ahsan Ejaz
- Division of Nephrology, Hypertension, and Transplantation, University of Florida, Gainesville, Florida
| | - Wataru Kitagawa
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - Gabriela E Garcia
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| | - David T Bonthron
- Leeds Institute of Biomedical & Clinical Sciences, University of Leeds, Leeds, United Kingdom; and
| | - Aruna Asipu
- Leeds Institute of Biomedical & Clinical Sciences, University of Leeds, Leeds, United Kingdom; and
| | - Christine P Diggle
- Leeds Institute of Biomedical & Clinical Sciences, University of Leeds, Leeds, United Kingdom; and
| | | | - Takahiko Nakagawa
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado; TMK Project, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Richard J Johnson
- The Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado, Denver, Colorado
| |
Collapse
|
486
|
Zelenchuk LV, Hedge AM, Rowe PSN. PHEX mimetic (SPR4-peptide) corrects and improves HYP and wild type mice energy-metabolism. PLoS One 2014; 9:e97326. [PMID: 24839967 PMCID: PMC4026222 DOI: 10.1371/journal.pone.0097326] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/17/2014] [Indexed: 12/19/2022] Open
Abstract
CONTEXT PHEX or DMP1 mutations cause hypophosphatemic-rickets and altered energy metabolism. PHEX binds to DMP1-ASARM-motif to form a complex with α5β3 integrin that suppresses FGF23 expression. ASARM-peptides increase FGF23 by disrupting the PHEX-DMP1-Integrin complex. We used a 4.2 kDa peptide (SPR4) that binds to ASARM-peptide/motif to study the DMP1-PHEX interaction and to assess SPR4 for the treatment of energy metabolism defects in HYP and potentially other bone-mineral disorders. DESIGN Subcutaneously transplanted osmotic pumps were used to infuse SPR4-peptide or vehicle (VE) into wild-type mice (WT) and HYP-mice (PHEX mutation) for 4 weeks. RESULTS SPR4 partially corrected HYP mice hypophosphatemia and increased serum 1.25(OH)2D3. Serum FGF23 remained high and PTH was unaffected. WT-SPR4 mice developed hypophosphatemia and hypercalcemia with increased PTH, FGF23 and 1.25(OH)2D3. SPR4 increased GAPDH HYP-bone expression 60× and corrected HYP-mice hyperglycemia and hypoinsulinemia. HYP-VE serum uric-acid (UA) levels were reduced and SPR4 infusion suppressed UA levels in WT-mice but not HYP-mice. SPR4 altered leptin, adiponectin, and sympathetic-tone and increased the fat mass/weight ratio for HYP and WT mice. Expression of perlipin-2 a gene involved in obesity was reduced in HYP-VE and WT-SPR4 mice but increased in HYP-SPR4 mice. Also, increased expression of two genes that inhibit insulin-signaling, ENPP1 and ESP, occurred with HYP-VE mice. In contrast, SPR4 reduced expression of both ENPP1 and ESP in WT mice and suppressed ENPP1 in HYP mice. Increased expression of FAM20C and sclerostin occurred with HYP-VE mice. SPR4 suppressed expression of FAM20C and sclerostin in HYP and WT mice. CONCLUSIONS ASARM peptides and motifs are physiological substrates for PHEX and modulate osteocyte PHEX-DMP1-α5β3-integrin interactions and thereby FGF23 expression. These interactions also provide a nexus that regulates bone and energy metabolism. SPR4 suppression of sclerostin and/or sequestration of ASARM-peptides improves energy metabolism and may have utility for treating familial rickets, osteoporosis, obesity and diabetes.
Collapse
Affiliation(s)
- Lesya V. Zelenchuk
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| | - Anne-Marie Hedge
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| | - Peter S. N. Rowe
- Internal Medicine, The Kidney Institute, Kansas University Medical Center (KUMC), Kansas City, Kansas, United States of America
| |
Collapse
|
487
|
Cicerchi C, Li N, Kratzer J, Garcia G, Roncal-Jimenez CA, Tanabe K, Hunter B, Rivard CJ, Sautin YY, Gaucher EA, Johnson RJ, Lanaspa MA. Uric acid-dependent inhibition of AMP kinase induces hepatic glucose production in diabetes and starvation: evolutionary implications of the uricase loss in hominids. FASEB J 2014; 28:3339-50. [PMID: 24755741 DOI: 10.1096/fj.13-243634] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
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.
Collapse
Affiliation(s)
- Christina Cicerchi
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Nanxing Li
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - James Kratzer
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA; and
| | - Gabriela Garcia
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Katsuyuki Tanabe
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Brandi Hunter
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Yuri Y Sautin
- School of Medicine, University of Florida, Gainesville, Florida, USA
| | - Eric A Gaucher
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA; and
| | - Richard J Johnson
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Miguel A Lanaspa
- School of Medicine, University of Colorado Denver, Aurora, Colorado, USA;
| |
Collapse
|
488
|
High-fat feeding increases hepatic vitamin C synthesis and its circulatory mobilization in mice. Eur J Nutr 2014; 53:1441-4. [DOI: 10.1007/s00394-014-0694-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/24/2014] [Indexed: 12/23/2022]
|
489
|
Evolutionary history and metabolic insights of ancient mammalian uricases. Proc Natl Acad Sci U S A 2014; 111:3763-8. [PMID: 24550457 DOI: 10.1073/pnas.1320393111] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
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.
Collapse
|
490
|
Shulga N, Pastorino JG. Mitoneet mediates TNFα-induced necroptosis promoted by exposure to fructose and ethanol. J Cell Sci 2013; 127:896-907. [PMID: 24357718 DOI: 10.1242/jcs.140764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Fructose and ethanol are metabolized principally in the liver and are both known to contribute to the development of hepatic steatosis that can progress to hepatic steatohepatitis. The present study indentifies a synergistic interaction between fructose and ethanol in promoting hepatocyte sensitivity to TNFα-induced necroptosis. Concurrent exposure to fructose and ethanol induces the overexpression of the CDGSH iron-sulfur domain-containing protein 1 (CISD1 or mitoneet), which is localized to the outer mitochondrial membrane. The increased expression of mitoneet primes the hepatocyte for TNFα-induced cytotoxicity. Treatment with TNFα induces the translocation of a Stat3-Grim-19 complex to the mitochondria, which binds to mitoneet and promotes the rapid release of its 2Fe-2S cluster, causing an accumulation of mitochondrial iron. The dramatic increase of mitochondrial iron provokes a surge in formation of reactive oxygen species, resulting in mitochondrial injury and cell death. Additionally, mitoneet is constitutively expressed at high levels in L929 fibrosarcoma cells and is required for L929 cells to undergo TNFα-induced necroptosis in the presence of caspase inhibition, indicating the importance of mitoneet to the necroptotic form of cell death.
Collapse
Affiliation(s)
- Nataly Shulga
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | | |
Collapse
|
491
|
Fructokinase activity mediates dehydration-induced renal injury. Kidney Int 2013; 86:294-302. [PMID: 24336030 PMCID: PMC4120672 DOI: 10.1038/ki.2013.492] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/20/2013] [Accepted: 10/03/2013] [Indexed: 12/28/2022]
Abstract
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.
Collapse
|
492
|
Quintero M, Gonzalez-Martin MDC, Vega-Agapito V, Gonzalez C, Obeso A, Farré R, Agapito T, Yubero S. The effects of intermittent hypoxia on redox status, NF-κB activation, and plasma lipid levels are dependent on the lowest oxygen saturation. Free Radic Biol Med 2013; 65:1143-1154. [PMID: 24002010 DOI: 10.1016/j.freeradbiomed.2013.08.180] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 08/01/2013] [Accepted: 08/23/2013] [Indexed: 12/11/2022]
Abstract
Obstructive sleep apnea syndrome (OSAS) is described as repetitive obstructions of the upper airways during sleep, causing concomitant episodes of systemic hypoxia and associated cardiovascular and metabolic pathologies. The mechanisms generating these pathologies are controversial. Because recurrent hypoxia is the element of inadequate respiration that leads to the pathology, experimental models of OSAS consist in the exposure of the animals to intermittent hypoxia (IH) by cycling O2 percentages in their habitats. A proposed mechanism linking the IH of OSAS to pathologies is the increased production of reactive oxygen species (ROS). However, it has been argued that many patients seem to lack oxidative stress and that, to augment ROS in IH animals, intense hypoxia, seldom encountered in patients, has to be applied. To solve the controversy, we have exposed rats to two intensities of IH (cycles of 10 or 5% O2, 40s, and then 21% O2, 80s; 8h/day, 15 days). We then measured reduced and oxidized glutathione and lipid peroxide levels, aconitase and fumarase activities, and ROS-disposal enzyme activity in liver, brain, and lung. Liver levels of nuclear NF-κB-p65 and plasma C-reactive protein (CRP), as well as lipid levels, were also assessed. Lowest hemoglobin saturations were 91.7 ± 0.8 and 73.5 ± 1.4%. IH caused tissue-specific oxidative stress related to hypoxic intensity. Nuclear NF-κB-p65 and lipid content in the liver and CRP in the plasma all increased with IH intensity, as did both plasma triglycerides and cholesterol. We conclude that IH, even of moderate intensity, causes oxidative stress probably related to the pathologies encountered in OSAS patients.
Collapse
Affiliation(s)
- Miguel Quintero
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - María Del Carmen Gonzalez-Martin
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Victoria Vega-Agapito
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Constancio Gonzalez
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain; CIBER Enfermedades Respiratorias, Institute of Salud Carlos III, 28029 Madrid, Spain
| | - Ana Obeso
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain; CIBER Enfermedades Respiratorias, Institute of Salud Carlos III, 28029 Madrid, Spain
| | - Ramon Farré
- CIBER Enfermedades Respiratorias, Institute of Salud Carlos III, 28029 Madrid, Spain; Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain
| | - Teresa Agapito
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain; CIBER Enfermedades Respiratorias, Institute of Salud Carlos III, 28029 Madrid, Spain
| | - Sara Yubero
- Institute of Molecular Biology and Genetics, CSIC, Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47005 Valladolid, Spain; CIBER Enfermedades Respiratorias, Institute of Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
493
|
Ishimoto T, Lanaspa MA, Rivard CJ, Roncal-Jimenez CA, Orlicky DJ, Cicerchi C, McMahan RH, Abdelmalek MF, Rosen HR, Jackman MR, MacLean PS, Diggle CP, Asipu A, Inaba S, Kosugi T, Sato W, Maruyama S, Sánchez-Lozada LG, Sautin YY, Hill JO, Bonthron DT, Johnson RJ. High-fat and high-sucrose (western) diet induces steatohepatitis that is dependent on fructokinase. Hepatology 2013; 58:1632-43. [PMID: 23813872 PMCID: PMC3894259 DOI: 10.1002/hep.26594] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 05/01/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
Abstract
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.
Collapse
Affiliation(s)
- Takuji Ishimoto
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Christopher J. Rivard
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Carlos A. Roncal-Jimenez
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - David J. Orlicky
- Department of Pathology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Christina Cicerchi
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Rachel H. McMahan
- Division of Gastroenterology and Hepatology, University of Colorado Denver, Denver, CO, 80045, USA
| | | | - Hugo R. Rosen
- Division of Gastroenterology and Hepatology, University of Colorado Denver, Denver, CO, 80045, USA
| | - Matthew R. Jackman
- Division of Endocrinology, Colorado Nutrition Obesity Research Center, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Paul S. MacLean
- Division of Endocrinology, Colorado Nutrition Obesity Research Center, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Christine P. Diggle
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK
| | - Aruna Asipu
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK
| | - Shinichiro Inaba
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Tomoki Kosugi
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Waichi Sato
- Departments of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Shoichi Maruyama
- Departments of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Laura G. Sánchez-Lozada
- Lab. of Renal Physiopathology & Dept. of Nephrology. INC Ignacio Chavez, Mexico City, 14080, Mexico
| | - Yuri Y. Sautin
- Division of Nephrology and Hypertension, University of Florida, Gainesville, FL, 32610, USA
| | - James O. Hill
- Anschutz Health and Wellness Center, University of Colorado Denver, Aurora, CO, 80045, USA
| | - David T. Bonthron
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, 80045, USA,Division of Nephrology and Hypertension, University of Florida, Gainesville, FL, 32610, USA,Address correspondence and reprint requests to Richard J Johnson, M.D.. Current address: University of Colorado Denver, Division of Renal Diseases and Hypertension, Box C281, 12700 E 19th Ave, Research 2 Room P15-7006, Aurora, CO, 80045, USA. Tel: 303 724 4898 Fax: 303 724 4831.
| |
Collapse
|
494
|
Lee D, Choi WJ, Oh JS, Yi MK, Han SW, Yun JW, Han SH. The relevance of hyperuricemia and metabolic syndrome and the effect of blood lead level on uric Acid concentration in steelmaking workers. Ann Occup Environ Med 2013; 25:27. [PMID: 24472219 PMCID: PMC3923343 DOI: 10.1186/2052-4374-25-27] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/13/2013] [Indexed: 02/07/2023] Open
Abstract
Objectives Uric acid concentration is known to increase the prevalence of metabolic syndrome by affecting its components, resulting in increased risk of cerebrovascular and cardiovascular diseases, and long-term lead exposure is known to affect this serum uric acid level. In this study, we aimed to examine the association between the causes of hyperuricemia and metabolic syndrome, and to determine whether an increased blood lead level affects hyperuricemia. Method Anthropometric measurements, surveys, and blood tests were conducted between May and June 2012 in 759 men working in the steelmaking process at a domestic steel company. Workers were divided into 2 groups according to the presence or absence of hyperuricemia, and an analysis was performed to examine its association with metabolic syndrome. In addition, the workers were divided into 3 groups according to the blood lead level to analyze the association between blood lead and hyperuricemia. Results The geometric mean (standard deviation) of the blood lead levels in the hyperuricemia group was significantly higher than that of the healthy group (3.8 [1.8] vs. 3.3 [1.8] μg/dL). The adjusted odds ratio for metabolic syndrome of the hyperuricemia group increased significantly to 1.787 (1.125–2.839) compared with the healthy group. In addition, the adjusted odds ratios for the occurrence of hyperuricemia in the tertile 2 (2.61–4.50 μg/dL) and tertile 3 groups (>4.50 μg/dL) according to blood lead level significantly increased to 1.763 (1.116–2.784) and 1.982 (1.254–3.132), respectively, compared with the tertile 1 group (< 2.61 μg/dL). Conclusion Hyperuricemia is believed to function as an independent risk factor for metabolic syndrome, while lead seems to increase the serum uric acid level even at a considerably low blood level. Therefore, attention should be given to patients with hyperuricemia and metabolic syndrome who are prone to lead exposure, and a prospective study should be conducted to identify their causal relationship.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sang-Hwan Han
- Department of Occupational & Environmental Medicine, Gachon University Gil Medical Center, Incheon, Korea.
| |
Collapse
|
495
|
He H, Wang S, Li X, Wang H, Zhang W, Yuan L, Liu X. A novel metabolic balance model for describing the metabolic disruption of and interactions between cardiovascular-related markers during acute myocardial infarction. Metabolism 2013; 62:1357-66. [PMID: 23702382 DOI: 10.1016/j.metabol.2013.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/09/2013] [Accepted: 04/13/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVE After acute myocardial infarction (AMI), an integral evaluation of risk using multimarker approach and the understanding of the pathophysiological processes involved have recently received much attention. This study aimed to develop a model to integrally evaluate the metabolic disruption of cardiovascular-related markers and unveil their interactions after AMI. METHODS AMI was induced in rats by coronary artery ligation. Several cardiovascular-related markers in plasma and the heart were determined during AMI. A metabolic balance model was developed using matrix equations to assess the metabolic disturbance of, and interactions between, these markers. RESULTS Metabolic balance maps intuitively depicted the metabolic disruption of cardiovascular-related markers after AMI. The deviation and magnitude of the disruption were quantitatively and integrally described by φ and k (the dynamic parameter of metabolic balance disruption), respectively. The metabolic balance was disturbed in both the circulatory system and the heart post-AMI. All of the measured markers appeared to be interactional. Among these markers, kidney function and dimethylarginine dimethylaminohydrolase (DDAH) activity in the heart showed a potent effect on the other markers, whereas asymmetric dimethylarginine (ADMA) levels in plasma and adenosine triphosphate (ATP) contents in the heart were susceptible to the effects of the other markers. CONCLUSION A metabolic balance model was developed to integrally evaluate the disruption of cardiovascular-related markers after AMI, which proposes a new method for evaluating the disease state post-AMI using a multimarker approach. The unveiled interactions between these cardiovascular-related markers are helpful in understanding the pathophysiological processes.
Collapse
Affiliation(s)
- Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | | | | | |
Collapse
|
496
|
Johnson RJ, Nakagawa T, Sanchez-Lozada LG, Shafiu M, Sundaram S, Le M, Ishimoto T, Sautin YY, Lanaspa MA. Sugar, uric acid, and the etiology of diabetes and obesity. Diabetes 2013; 62:3307-15. [PMID: 24065788 PMCID: PMC3781481 DOI: 10.2337/db12-1814] [Citation(s) in RCA: 494] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intake of added sugars, such as from table sugar (sucrose) and high-fructose corn syrup has increased dramatically in the last hundred years and correlates closely with the rise in obesity, metabolic syndrome, and diabetes. Fructose is a major component of added sugars and is distinct from other sugars in its ability to cause intracellular ATP depletion, nucleotide turnover, and the generation of uric acid. In this article, we revisit the hypothesis that it is this unique aspect of fructose metabolism that accounts for why fructose intake increases the risk for metabolic syndrome. Recent studies show that fructose-induced uric acid generation causes mitochondrial oxidative stress that stimulates fat accumulation independent of excessive caloric intake. These studies challenge the long-standing dogma that "a calorie is just a calorie" and suggest that the metabolic effects of food may matter as much as its energy content. The discovery that fructose-mediated generation of uric acid may have a causal role in diabetes and obesity provides new insights into pathogenesis and therapies for this important disease.
Collapse
Affiliation(s)
- Richard J. Johnson
- Division of Kidney Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
- Division of Nephrology, Eastern Colorado Health Care System, Department of Veteran Affairs, Denver, Colorado
- Corresponding author: Richard J. Johnson,
| | - Takahiko Nakagawa
- Division of Kidney Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
- TMK Project, Medical Innovation Center, Kyoto University, Kyoto, Japan
| | - L. Gabriela Sanchez-Lozada
- Laboratory of Renal Physiopathology and Department of Nephrology, Instituto Nacional de Cardiologia I.Ch., Mexico City, Mexico
| | | | - Shikha Sundaram
- Division of Pediatric Gastroenterology, Children’s Hospital, Aurora, Colorado
| | - Myphuong Le
- Division of Kidney Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Takuji Ishimoto
- Division of Kidney Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Yuri Y. Sautin
- Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, Florida
| | - Miguel A. Lanaspa
- Division of Kidney Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
497
|
Mastrocola R, Collino M, Rogazzo M, Medana C, Nigro D, Boccuzzi G, Aragno M. Advanced glycation end products promote hepatosteatosis by interfering with SCAP-SREBP pathway in fructose-drinking mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G398-407. [PMID: 23868406 DOI: 10.1152/ajpgi.00450.2012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clinical studies have linked the increased consumption of fructose to the development of obesity, dyslipidemia, and impaired glucose tolerance, and a role in hepatosteatosis development is presumed. Fructose can undergo a nonenzymatic reaction from which advanced glycation end products (AGEs) are derived, leading to the formation of dysfunctional, fructosylated proteins; however, the in vivo formation of AGEs from fructose is still less known than that from glucose. In the present study C57Bl/6J mice received 15% (wt/vol) fructose (FRT) or 15% (wt/vol) glucose (GLC) in water to drink for 30 wk, resembling human habit to consume sugary drinks. At the end of the protocol both FRT- and GLC-drinking mice had increased fasting glycemia, glucose intolerance, altered plasma lipid profile, and marked hepatosteatosis. FRT mice had higher hepatic triglycerides deposition than GLC, paralleled by a greater increased expression and activity of the sterol regulatory element-binding protein 1 (SREBP1), the transcription factor responsible for the de novo lipogenesis, and of its activating protein SCAP. LC-MS analysis showed a different pattern of AGE production in liver tissue between FRT and GLC mice, with larger amount of carboxymethyl lysine (CML) generated by fructose. Double immunofluorescence and coimmunoprecipitation analysis revealed an interaction between CML and SCAP that could lead to prolonged activation of SREBP1. Overall, the high levels of CML and activation of SCAP/SREBP pathway associated to high fructose exposure here reported may suggest a key role of this signaling pathway in mediating fructose-induced lipogenesis.
Collapse
Affiliation(s)
- Raffaella Mastrocola
- Dept. of Clinical and Biological Sciences, Experimental Medicine and Clinical Pathology Unit, Univ. of Turin, C.so Raffaello 30, 10125 Torino, Italy.
| | | | | | | | | | | | | |
Collapse
|
498
|
Stenvinkel P, Fröbert O, Anderstam B, Palm F, Eriksson M, Bragfors-Helin AC, Qureshi AR, Larsson T, Friebe A, Zedrosser A, Josefsson J, Svensson M, Sahdo B, Bankir L, Johnson RJ. Metabolic changes in summer active and anuric hibernating free-ranging brown bears (Ursus arctos). PLoS One 2013; 8:e72934. [PMID: 24039826 PMCID: PMC3767665 DOI: 10.1371/journal.pone.0072934] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/12/2013] [Indexed: 12/31/2022] Open
Abstract
The brown bear (Ursus arctos) hibernates for 5 to 6 months each winter and during this time ingests no food or water and remains anuric and inactive. Despite these extreme conditions, bears do not develop azotemia and preserve their muscle and bone strength. To date most renal studies have been limited to small numbers of bears, often in captive environments. Sixteen free-ranging bears were darted and had blood drawn both during hibernation in winter and summer. Samples were collected for measurement of creatinine and urea, markers of inflammation, the calcium-phosphate axis, and nutritional parameters including amino acids. In winter the bear serum creatinine increased 2.5 fold despite a 2-fold decrease in urea, indicating a remarkable ability to recycle urea nitrogen during hibernation. During hibernation serum calcium remained constant despite a decrease in serum phosphate and a rise in FGF23 levels. Despite prolonged inactivity and reduced renal function, inflammation does not ensue and bears seem to have enhanced antioxidant defense mechanisms during hibernation. Nutrition parameters showed high fat stores, preserved amino acids and mild hyperglycemia during hibernation. While total, essential, non-essential and branched chain amino acids concentrations do not change during hibernation anorexia, changes in individual amino acids ornithine, citrulline and arginine indicate an active, although reduced urea cycle and nitrogen recycling to proteins. Serum uric acid and serum fructose levels were elevated in summer and changes between seasons were positively correlated. Further studies to understand how bears can prevent the development of uremia despite minimal renal function during hibernation could provide new therapeutic avenues for the treatment of human kidney disease.
Collapse
Affiliation(s)
- Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Ole Fröbert
- Department of Cardiology, University Hospital of Örebro, Örebro, Sweden
| | - Björn Anderstam
- Division of Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Palm
- Department of Medical & Health Sciences, Experimental Renal Medicine, Linköping University, Linköping, Sweden
| | - Monica Eriksson
- Division of Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ann-Christin Bragfors-Helin
- Division of Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Division of Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tobias Larsson
- Division of Renal Medicine, Department of Clinical Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Friebe
- Zoologisches Institut, Goethe-Universität, Frankfurt am Main, Germany
| | - Andreas Zedrosser
- Faculty of Arts & Sciences, Department of Environmental & Health Studies, Telemark University College, Porsgrunn, Norway
- Institute for Wildlife Biology & Game Management, University for Natural Research & Life Sciences, Vienna, Austria
| | - Johan Josefsson
- Department of Cardiology, University Hospital of Örebro, Örebro, Sweden
| | - My Svensson
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | - Berolla Sahdo
- Department of Clinical Medicine, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Lise Bankir
- INSERM Unit 872-E2, Centre de Recherche des Cordeliers, Paris, France
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Denver, Colorado, United States of America
| |
Collapse
|
499
|
Abstract
The inexorable increase in the prevalence of obesity is a global health concern, which will result in a concomitant escalation in health-care costs. Obesity-related metabolic syndrome affects approximately 25% of adults and is associated with cardiovascular and renal disease. The heart and kidneys are physiologically interdependent, and the pathological effects of obesity can lead to cardiorenal syndrome and, ultimately, kidney and heart failure. Weight loss can prevent or ameliorate obesity-related cardiorenal syndrome, but long-term maintenance of a healthy weight has been difficult to achieve through lifestyle changes or pharmacotherapy. Bariatric surgery offers both sustained weight loss and favourable metabolic changes, including dramatic improvements in glycaemic control and symptoms of type 2 diabetes mellitus. Procedures such as Roux-en-Y gastric bypass offer immediate multisystemic benefits, including bile flow alteration, reduced gastric size, anatomical gut rearrangement and altered flow of nutrients, vagal manipulation and enteric hormone modulation. In patients with cardiorenal syndrome, bariatric surgery also offers renoprotection and cardioprotection, and attenuates both kidney and heart failure by improving organ perfusion and reversing metabolic dysfunction. However, further research is required to understand how bariatric surgery acts on the cardiorenal axis, and its pioneering role in novel treatments and interventions for cardiorenal disease.
Collapse
|
500
|
Abstract
PURPOSE OF REVIEW Carbohydrate consumption has been implicated in the metabolic syndrome and nonalcoholic fatty liver disease (NAFLD). Reviewed here is basis for this relationship and the recent additional evidence that excessive dietary carbohydrate consumption, especially excessive fructose or sucrose consumption, is playing a role in the epidemic of NAFLD. RECENT FINDINGS A recent cross-sectional epidemiological study has linked fructose consumption to the severity of fibrosis in patients with NAFLD. Clinical trials have shown that consumption of fructose-containing beverages, either has fructose or sucrose, contribute to the development of NAFLD compared to isocaloric alternatives, and that genetic polymorphisms that increase the entry of glucose into lipogenic pathways are associated with fatty liver disease. New animal studies provide additional evidence on the role of carbohydrate-induced de-novo lipogenesis and the gut microbiome in fructose-induced NAFLD. Data also suggest that fructose-induced uric acid production in the liver also plays a role in NAFLD independent of the role of fructose as a substrate for lipogenesis. SUMMARY Epidemiological studies, clinical trials, and animal studies continue to point to excess dietary carbohydrate, and especially fructose, in contributing to the risk factors for NAFLD.
Collapse
|