1
|
Chen L, Xu T, Wang J, Wang Z, Pan Y, Kong L. Siwu tablet attenuates high fructose-induced glomerular podocyte senescence in rats through increasing Nup155 to promote INO80 mRNA nuclear export. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118878. [PMID: 39362331 DOI: 10.1016/j.jep.2024.118878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/23/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Siwu tablet (SWT), derived from a traditional Chinese medicinal formula named Siwu decoction, is widely used for blood deficiency syndrome. Siwu decoction and its derived formulas have been proven to improve renal anemia and prevent senescence. Whether SWT prevents glomerular podocyte senescence and the underlying molecular mechanism remains unknow. AIM OF THE STUDY To elucidate the protective effect and possible mechanism of SWT on glomerular podocyte senescence. MATERIAL AND METHODS Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was performed to characterize components of SWT. Male Sprague-Dawley rats were given 10% fructose drinking water for 16 weeks. SWT (810 and 1620 mg/kg) was administered orally for the last 8 weeks. The assays of senescence-associated beta-galactosidase (SA-β-gal) staining, immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot as well as enzyme linked immunosorbent assay were performed to evaluate rat glomerular podocyte senescence. The mRNA and protein levels of nucleoporin 155 (Nup155) and inositol requiring mutant 80 (INO80) in rat glomeruli were detected by qRT-PCR, Western blot and immunofluorescence. Foot processes and nuclear pore complexes (NPCs) of rat glomerular podocytes were visualized by transmission electron microscopy. RESULTS One hundred and fifty-nine components were preliminarily identified in SWT. The results of animal experiments showed that SWT decreased the activity of SA-β-gal, protein levels of p16, p21, p53 and phosphorylated histone H2AX (γ-H2AX), and mRNA levels of interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α) in glomeruli of high fructose-fed rats. As expected, SWT increased renal cortex erythropoietin mRNA expression and serum erythropoietin concentration in this animal model. SWT reduced urine albumin-to-creatinine ratio and serum levels of uric acid, creatinine and blood urea nitrogen, and recovered glomerular structure injury in high fructose-fed rats. It up-regulated mRNA and protein levels of Nup155 and the number of podocyte NPCs, and subsequently reinforced mRNA nuclear export and protein expression of INO80 in rat glomeruli under high fructose stimulation. CONCLUSIONS SWT ameliorates glomerular podocyte senescence in high fructose-fed rats possibly by increasing Nup155 to promote INO80 mRNA nuclear export.
Collapse
Affiliation(s)
- Li Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Tangdi Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Jiahao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Zixuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Ying Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing, PR China.
| |
Collapse
|
2
|
Le Page AK, Johnson EC, Greenberg JH. Is mild dehydration a risk for progression of childhood chronic kidney disease? Pediatr Nephrol 2024; 39:3177-3191. [PMID: 38632124 DOI: 10.1007/s00467-024-06332-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 04/19/2024]
Abstract
Children with chronic kidney disease (CKD) can have an inherent vulnerability to dehydration. Younger children are unable to freely access water, and CKD aetiology and stage can associate with reduced kidney concentrating capacity, which can also impact risk. This article aims to review the risk factors and consequences of mild dehydration and underhydration in CKD, with a particular focus on evidence for risk of CKD progression. We discuss that assessment of dehydration in the CKD population is more challenging than in the healthy population, thus complicating the definition of adequate hydration and clinical research in this field. We review pathophysiologic studies that suggest mild dehydration and underhydration may cause hyperfiltration injury and impact renal function, with arginine vasopressin as a key mediator. Randomised controlled trials in adults have not shown an impact of improved hydration in CKD outcomes, but more vulnerable populations with baseline low fluid intake or poor kidney concentrating capacity need to be studied. There is little published data on the frequency of dehydration, and risk of complications, acute or chronic, in children with CKD. Despite conflicting evidence and the need for more research, we propose that paediatric CKD management should routinely include an assessment of individual dehydration risk along with a treatment plan, and we provide a framework that could be used in outpatient settings.
Collapse
Affiliation(s)
- Amelia K Le Page
- Department of Nephrology, Monash Children's Hospital, Clayton, VIC, Australia.
- Department of Pediatrics, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
| | - Evan C Johnson
- Division of Kinesiology & Health, College of Health Sciences, University of Wyoming, Laramie, WY, USA
| | - Jason H Greenberg
- Section of Nephrology, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Clinical and Translational Research Accelerator, Yale University, New Haven, CT, USA
| |
Collapse
|
3
|
Baharuddin B. The Impact of Fructose Consumption on Human Health: Effects on Obesity, Hyperglycemia, Diabetes, Uric Acid, and Oxidative Stress With a Focus on the Liver. Cureus 2024; 16:e70095. [PMID: 39355469 PMCID: PMC11444807 DOI: 10.7759/cureus.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
Excessive fructose consumption, primarily through processed foods and beverages, has become a significant public health concern due to its association with various metabolic disorders. This review examines the impact of fructose on human health, focusing on its role in obesity, insulin resistance, hyperglycemia, type 2 diabetes, uric acid production, and oxidative stress. Fructose metabolism, distinct from glucose, predominantly occurs in the liver, where it bypasses normal insulin regulation, leading to increased fat synthesis through de novo lipogenesis. This process contributes to the development of non-alcoholic fatty liver disease and elevates the risk of cardiovascular disease. Furthermore, fructose-induced adenosine triphosphate depletion activates purine degradation, increasing uric acid levels and exacerbating hyperuricemia. The overproduction of reactive oxygen species during fructose metabolism also drives oxidative stress, promoting inflammation and cellular damage. By synthesizing recent findings, this review underscores the importance of regulating fructose intake, implementing public health policies, and adopting lifestyle changes to mitigate these adverse effects.
Collapse
|
4
|
Chen L, Xu T, Wang Z, Wang C, Fang L, Kong L. Loss of Nup155 promotes high fructose-driven podocyte senescence by inhibiting INO80 mRNA nuclear export. J Adv Res 2024:S2090-1232(24)00329-1. [PMID: 39111625 DOI: 10.1016/j.jare.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/11/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024] Open
Abstract
INTRODUCTION Podocyte senescence causes podocyte loss and glomerulopathy. Excessive fructose intake is a risk factor for podocyte injury. However, whether high fructose promotes podocyte senescence remains unknown. OBJECTIVES To explore the pathological mechanism by which high fructose drives podocyte senescence and find natural compounds to alleviate podocyte senescence. METHODS Podocyte senescence was characterized with senescence-associated beta-galactosidase (SA-β-gal) staining, Western blot, real-time quantitative polymerase chain reaction (qRT-PCR), comet assay and immunofluorescence. Proteomics analysis was performed to identify differentially expressed proteins in high fructose-exposed podocytes. Podocyte nuclear pore complexes (NPCs) and foot processes were observed by transmission electron microscopy. The mRNA and protein levels of nucleoporin 155 (Nup155) and inositol requiring mutant 80 (INO80) were detected by qRT-PCR, Western blot and immunofluorescence. Virtual screening was conducted to find natural compounds that target Nup155. RESULTS High fructose increased SA-β-gal activity, protein level of p53, p21, p16 and phosphorylated histone H2AX (γ-H2AX), as well as mRNA expression of interleukin-1β (IL-1β), IL-6 and tumor necrosis factor α (TNF-α) in rat glomeruli and podocytes. Proteomic analysis unraveled a crucial molecule Nup155, which was decreased in high fructose-induced podocyte senescence. Meanwhile, the number of podocyte NPCs was also decreased in vivo and in vitro. Consistently, high fructose suppressed nuclear export of INO80 mRNA, thereby down-regulated INO80 protein expression in podocyte senescence. Deletion of Nup155 inhibited INO80 mRNA nuclear export to induce podocyte senescence, whereas overexpression of Nup155 or INO80 alleviated high fructose-induced podocyte senescence. Ferulic acid was found to up-regulate Nup155 by both direct binding to stabilize Nup155 protein and enhancing its transcription, to promote INO80 mRNA nuclear export in the mitigation of high fructose-caused podocyte senescence. CONCLUSION High fructose induces podocyte senescence by decreasing Nup155 to inhibit INO80 mRNA nuclear export. Ferulic acid targeting Nup155 may be a potential strategy to prevent high fructose-induced podocyte senescence.
Collapse
Affiliation(s)
- Li Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Tangdi Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zixuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Chengzhi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China; Chemistry and Biomedicine Innovation Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China; Chemistry and Biomedicine Innovation Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China.
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
5
|
Zhang G, Li Z, Zhang S, Bai L, Zhou H, Zhang D. Anti-Type II Diabetic Effects of Coix Seed Prolamin Hydrolysates: Physiological and Transcriptomic Analyses. Foods 2024; 13:2203. [PMID: 39063287 PMCID: PMC11275950 DOI: 10.3390/foods13142203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Previous studies have demonstrated that enzymatically prepared coix seed prolamin hydrolysates (CHPs) contain several bioactive peptides that efficiently inhibit the activity of target enzymes (α-glucosidase and dipeptidyl kinase-IV) in type 2 diabetes mellitus (T2DM). However, the anti-T2DM effects and potential mechanisms of CHPs as a whole in vivo have not yet been systematically explored. Therefore, we evaluated the preventive, therapeutic, and modifying effects of CHPs on T2DM by combining physiological and liver transcriptomics with a T2DM mouse model. The results showed that sustained high-fructose intake led to prediabetic symptoms in mice, with abnormal fluctuations in blood glucose and blood lipid levels. Intervention with CPHs effectively prevented weight loss; regulated abnormal changes in blood glucose; improved impaired glucose tolerance; inhibited the abnormal expression of total cholesterol, triglycerides, and low-density lipoproteins; alleviated insulin resistance; and restored pancreatic islet tissue function in mice fed a high-fructose diet. In addition, we found that CHPs also play a palliative role in the loss of liver function and protect various organ tissues (including the liver, kidneys, pancreas, and heart), and are effective in preventing damage to the liver and pancreatic islet cells. We also found that the intake of CHPs reversed the abnormally altered hepatic gene profile in model mice and identified 381 differentially expressed genes that could serve as key genes for preventing the development of T2DM, which are highly correlated with multiple glycolipid metabolic pathways. We demonstrated that CHPs play a positive role in the normal functioning of the insulin signalling pathway dominated by the IRS-1/PI3K/AKT (insulin receptor substrates-1/phosphoinositide 3-kinase/protein kinase B) pathway. In summary, CHPs can be used as effective food-borne glucose-modifying components of healthy foods.
Collapse
Affiliation(s)
- Guifang Zhang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.Z.); (Z.L.); (S.Z.); (L.B.); (H.Z.)
- Food College, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Zhiming Li
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.Z.); (Z.L.); (S.Z.); (L.B.); (H.Z.)
- Food College, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shu Zhang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.Z.); (Z.L.); (S.Z.); (L.B.); (H.Z.)
- Food College, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Lu Bai
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.Z.); (Z.L.); (S.Z.); (L.B.); (H.Z.)
- Food College, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Hangqing Zhou
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.Z.); (Z.L.); (S.Z.); (L.B.); (H.Z.)
- Food College, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Dongjie Zhang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (G.Z.); (Z.L.); (S.Z.); (L.B.); (H.Z.)
- Food College, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, China
| |
Collapse
|
6
|
Korsmo HW, Ekperikpe US, Daehn IS. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:712. [PMID: 38929151 PMCID: PMC11200862 DOI: 10.3390/antiox13060712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.
Collapse
Affiliation(s)
| | | | - Ilse S. Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1243, New York, NY 10029, USA
| |
Collapse
|
7
|
Chen DQ, Han J, Liu H, Feng K, Li P. Targeting pyruvate kinase M2 for the treatment of kidney disease. Front Pharmacol 2024; 15:1376252. [PMID: 38910890 PMCID: PMC11190346 DOI: 10.3389/fphar.2024.1376252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/05/2024] [Indexed: 06/25/2024] Open
Abstract
Pyruvate kinase M2 (PKM2), a rate limiting enzyme in glycolysis, is a cellular regulator that has received extensive attention and regards as a metabolic regulator of cellular metabolism and energy. Kidney is a highly metabolically active organ, and glycolysis is the important energy resource for kidney. The accumulated evidences indicates that the enzymatic activity of PKM2 is disturbed in kidney disease progression and treatment, especially diabetic kidney disease and acute kidney injury. Modulating PKM2 post-translational modification determines its enzymatic activity and nuclear translocation that serves as an important interventional approach to regulate PKM2. Emerging evidences show that PKM2 and its post-translational modification participate in kidney disease progression and treatment through modulating metabolism regulation, podocyte injury, fibroblast activation and proliferation, macrophage polarization, and T cell regulation. Interestingly, PKM2 activators (TEPP-46, DASA-58, mitapivat, and TP-1454) and PKM2 inhibitors (shikonin, alkannin, compound 3k and compound 3h) have exhibited potential therapeutic property in kidney disease, which indicates the pleiotropic effects of PKM2 in kidney. In the future, the deep investigation of PKM2 pleiotropic effects in kidney is urgently needed to determine the therapeutic effect of PKM2 activator/inhibitor to benefit patients. The information in this review highlights that PKM2 functions as a potential biomarker and therapeutic target for kidney diseases.
Collapse
Affiliation(s)
- Dan-Qian Chen
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Jin Han
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Xi’an Chang’an District Hospital, Xi’an, Shaanxi, China
| | - Hui Liu
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Kai Feng
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
8
|
Wang X, Chang HC, Gu X, Han W, Mao S, Lu L, Jiang S, Ding H, Han S, Qu X, Bao Z. Renal lipid accumulation and aging linked to tubular cells injury via ANGPTL4. Mech Ageing Dev 2024; 219:111932. [PMID: 38580082 DOI: 10.1016/j.mad.2024.111932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Renal tubular epithelial cells are vulnerable to stress-induced damage, including excessive lipid accumulation and aging, with ANGPTL4 potentially playing a crucial bridging role between these factors. In this study, RNA-sequencing was used to identify a marked increase in ANGPTL4 expression in kidneys of diet-induced obese and aging mice. Overexpression and knockout of ANGPTL4 in renal tubular epithelial cells (HK-2) was used to investigate the underlying mechanism. Subsequently, ANGPTL4 expression in plasma and kidney tissues of normal young controls and elderly individuals was analyzed using ELISA and immunohistochemical techniques. RNA sequencing results showed that ANGPTL4 expression was significantly upregulated in the kidney tissue of diet-induced obesity and aging mice. In vitro experiments demonstrated that overexpression of ANGPTL4 in HK-2 cells led to increased lipid deposition and senescence. Conversely, the absence of ANGPTL4 appears to alleviate the impact of free fatty acids (FFA) on aging in HK-2 cells. Additionally, aging HK-2 cells exhibited elevated ANGPTL4 expression, and stress response markers associated with cell cycle arrest. Furthermore, our clinical evidence revealed dysregulation of ANGPTL4 expression in serum and kidney tissue samples obtained from elderly individuals compared to young subjects. Our study findings indicate a potential association between ANGPTL4 and age-related metabolic disorders, as well as injury to renal tubular epithelial cells. This suggests that targeting ANGPTL4 could be a viable strategy for the clinical treatment of renal aging.
Collapse
Affiliation(s)
- Xiaojun Wang
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Hung-Chen Chang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Xuchao Gu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Wanlin Han
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Shihang Mao
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, China
| | - Lili Lu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Shuai Jiang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Haiyong Ding
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Urologic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Shisheng Han
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xinkai Qu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| |
Collapse
|
9
|
Tsuruta H, Yasuda-Yamahara M, Yoshibayashi M, Kuwagata S, Yamahara K, Tanaka-Sasaki Y, Chin-Kanasaki M, Matsumoto S, Ema M, Kume S. Fructose overconsumption accelerates renal dysfunction with aberrant glomerular endothelial-mesangial cell interactions in db/db mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167074. [PMID: 38354758 DOI: 10.1016/j.bbadis.2024.167074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
For the advancement of DKD treatment, identifying unrecognized residual risk factors is essential. We explored the impact of obesity diversity derived from different carbohydrate qualities, with an emphasis on the increasing trend of excessive fructose consumption and its effect on DKD progression. In this study, we utilized db/db mice to establish a novel diabetic model characterized by fructose overconsumption, aiming to uncover the underlying mechanisms of renal damage. Compared to the control diet group, the fructose-fed db/db mice exhibited more pronounced obesity yet demonstrated milder glucose intolerance. Plasma cystatin C levels were elevated in the fructose model compared to the control, and this elevation was accompanied by enhanced glomerular sclerosis, even though albuminuria levels and tubular lesions were comparable. Single-cell RNA sequencing of the whole kidney highlighted an increase in Lrg1 in glomerular endothelial cells (GECs) in the fructose model, which appeared to drive mesangial fibrosis through enhanced TGF-β1 signaling. Our findings suggest that excessive fructose intake exacerbates diabetic kidney disease progression, mediated by aberrant Lrg1-driven crosstalk between GECs and mesangial cells.
Collapse
Affiliation(s)
- Hiroaki Tsuruta
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Mako Yasuda-Yamahara
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Mamoru Yoshibayashi
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Shogo Kuwagata
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Kosuke Yamahara
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Yuki Tanaka-Sasaki
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Masami Chin-Kanasaki
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu, Shiga, Japan.
| |
Collapse
|
10
|
Tero-Vescan A, Ștefănescu R, Istrate TI, Pușcaș A. Fructose-induced hyperuricaemia - protection factor or oxidative stress promoter? Nat Prod Res 2024:1-13. [PMID: 38522076 DOI: 10.1080/14786419.2024.2327624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/28/2024] [Indexed: 03/26/2024]
Abstract
Accumulating evidence suggests that dietary fructose may play a role in the hyperuricaemia development, but the precise mechanism remains unclear. Hyperuricaemia is characterised by excessive production and deposition of urate crystals, and the metabolism of fructose has been implicated in the elevation of serum urate levels. The association between fructose intake and the risk of hyperuricaemia is explained by the metabolism of fructose in the liver, small intestine, and kidney. Many studies have confirmed the correlation between fructose consumption and an increased risk of developing hyperuricaemia, but more prospective studies to fully elucidate the role of fructose intake in the pathogenesis of hyperuricaemia are needed. It is important to note that maintaining a balanced diet, and lifestyle is crucial when considering fructose intake. Limiting the consumption of products high in added sugars and maintaining a healthy weight can contribute to reducing the risk of hyperuricaemia and associated health complications.
Collapse
Affiliation(s)
- Amelia Tero-Vescan
- Medical Chemistry and Biochemistry Department, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Hamburg, Germany
| | - Ruxandra Ștefănescu
- Department of Pharmacognosy and Phytotherapy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Târgu Mures, Romania
| | - Tudor-Ionuț Istrate
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, Târgu Mures, Romania
| | - Amalia Pușcaș
- Biochemistry and Chemistry of the Environmental Factors Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, Târgu Mures, Romania
| |
Collapse
|
11
|
Chen Y, Liu Q, Meng X, Zhao L, Zheng X, Feng W. Catalpol ameliorates fructose-induced renal inflammation by inhibiting TLR4/MyD88 signaling and uric acid reabsorption. Eur J Pharmacol 2024; 967:176356. [PMID: 38325797 DOI: 10.1016/j.ejphar.2024.176356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/15/2023] [Accepted: 01/23/2024] [Indexed: 02/09/2024]
Abstract
Accumulating evidence suggests that excess fructose uptake induces metabolic syndrome and kidney injury. Here, we primarily investigated the influence of catalpol on fructose-induced renal inflammation in mice and explored its potential mechanism. Treatment with catalpol improved insulin sensitivity and hyperuricemia in fructose-fed mice. Hyperuricemia induced by high-fructose diet was associated with increases in the expressions of urate reabsorptive transporter URAT1 and GLUT9. Treatment with catalpol decreased the expressions of URAT1 and GLUT9. Futhermore, treatment with catalpol ameliorated renal inflammatory cell infiltration and podocyte injury, and these beneficial effects were associated with inhibiting the production of inflammatory cytokines including IL-1β, IL-18, IL-6 and TNF-α. Moreover, fructose-induced uric acid triggers an inflammatory response by activiting NLRP3 inflammasome, which then processes pro-inflammatory cytokines. Treatment with catalpol could inhibit the activation of NLRP3 inflammasome as well. Additionally, TLR4/MyD88 signaling was activated in fructose-fed mice, while treatment with catalpol inhibited this activation along with promoting NF-κB nuclear translocation in fructose-fed mice. Thus, our study demonstrated that catalpol could ameliorate renal inflammation in fructose-fed mice, attributing its beneficial effects to promoting uric acid excretion and inhibit the activation of TLR4/MyD88 signaling.
Collapse
Affiliation(s)
- Yan Chen
- College of Pharmacy, Henan University of Chinese Medicine, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, China
| | - Qingpu Liu
- College of Pharmacy, Henan University of Chinese Medicine, China; The Engineering and Technology Research Center of Quality Control and Evaluation for Chinese Medicine Development of Henan Province, China
| | - Xinyu Meng
- College of Pharmacy, Henan University of Chinese Medicine, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, China
| | - Liqin Zhao
- College of Pharmacy, Henan University of Chinese Medicine, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, China.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, China.
| |
Collapse
|
12
|
Menyhárt O, Győrffy B. Dietary approaches for exploiting metabolic vulnerabilities in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189062. [PMID: 38158024 DOI: 10.1016/j.bbcan.2023.189062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Renewed interest in tumor metabolism sparked an enthusiasm for dietary interventions to prevent and treat cancer. Changes in diet impact circulating nutrient levels in the plasma and the tumor microenvironment, and preclinical studies suggest that dietary approaches, including caloric and nutrient restrictions, can modulate tumor initiation, progression, and metastasis. Cancers are heterogeneous in their metabolic dependencies and preferred energy sources and can be addicted to glucose, fructose, amino acids, or lipids for survival and growth. This dependence is influenced by tumor type, anatomical location, tissue of origin, aberrant signaling, and the microenvironment. This review summarizes nutrient dependencies and the related signaling pathway activations that provide targets for nutritional interventions. We examine popular dietary approaches used as adjuvants to anticancer therapies, encompassing caloric restrictions, including time-restricted feeding, intermittent fasting, fasting-mimicking diets (FMDs), and nutrient restrictions, notably the ketogenic diet. Despite promising results, much of the knowledge on dietary restrictions comes from in vitro and animal studies, which may not accurately reflect real-life situations. Further research is needed to determine the optimal duration, timing, safety, and efficacy of dietary restrictions for different cancers and treatments. In addition, well-designed human trials are necessary to establish the link between specific metabolic vulnerabilities and targeted dietary interventions. However, low patient compliance in clinical trials remains a significant challenge.
Collapse
Affiliation(s)
- Otília Menyhárt
- Semmelweis University, Department of Bioinformatics, Tűzoltó u. 7-9, H-1094 Budapest, Hungary; Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Balázs Győrffy
- Semmelweis University, Department of Bioinformatics, Tűzoltó u. 7-9, H-1094 Budapest, Hungary; Research Centre for Natural Sciences, Cancer Biomarker Research Group, Institute of Enzymology, Magyar tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117 Budapest, Hungary.
| |
Collapse
|
13
|
Huang L, Zhang L, Zhang Z, Tan F, Ma Y, Zeng X, Cao D, Deng L, Liu Q, Sun H, Shen B, Liao X. Loss of nephric augmenter of liver regeneration facilitates acute kidney injury via ACSL4-mediated ferroptosis. J Cell Mol Med 2024; 28:e18076. [PMID: 38088220 PMCID: PMC10844764 DOI: 10.1111/jcmm.18076] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/01/2023] [Accepted: 11/25/2023] [Indexed: 02/08/2024] Open
Abstract
Ferroptosis, characterized by lipid accumulation in intracellular compartments, is related to acute kidney injury (AKI), but the mechanism remains obscure. In our previous study, the protective effect of augmenter of liver regeneration (ALR) on AKI was not fully clarified. In this study, we established an AKI mouse model by knocking out proximal tubule-specific ALR and an AKI cell model by inducing hypoxia, as well as enrolled AKI patients, to investigate the effects of ALR on ferroptosis and the progression of AKI. We found that ALR knockout aggravated ferroptosis and increased ROS accumulation and mitochondrial damage, whereas ALR overexpression attenuated ferroptosis through clearance of ROS and maintenance of mitochondrial morphology. Mechanistically, we demonstrated that ALR could directly bind to long-chain-fatty-acid-CoA ligase 4 (ACSL4) and further inhibit the expression of ACSL4 by interacting with certain regions. By resolution liquid chromatography coupled with triple quadruple mass spectrometry, we found that ALR could reduce the contents of polyunsaturated fatty acids, especially arachidonic acid. In addition, we showed that ALR binds to ACSL4 and attenuates oxylipin accumulation, exerting a protective effect against ferroptosis in AKI. Therefore, targeting renal ALR can attenuate ferroptosis and can offer a promising strategy for the treatment of AKI.
Collapse
Affiliation(s)
- Lili Huang
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Ling Zhang
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Zheng Zhang
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
- Department of Cell Biology and GeneticsChongqing Medical UniversityChongqingChina
| | - Fangyan Tan
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Yixin Ma
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xujia Zeng
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Dan Cao
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Lili Deng
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Qi Liu
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral HepatitisThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Hang Sun
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral HepatitisThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Bingbing Shen
- Department of NephrologyChongqing University Central Hospital, Chongqing Emergency Medical CenterChongqingChina
| | - Xiaohui Liao
- Department of NephrologyThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
- Kuanren Laboratory of Translational Lipidology, Centre for Lipid ResearchThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
14
|
Butcko AJ, Putman AK, Mottillo EP. The Intersection of Genetic Factors, Aberrant Nutrient Metabolism and Oxidative Stress in the Progression of Cardiometabolic Disease. Antioxidants (Basel) 2024; 13:87. [PMID: 38247511 PMCID: PMC10812494 DOI: 10.3390/antiox13010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/06/2023] [Accepted: 01/07/2024] [Indexed: 01/23/2024] Open
Abstract
Cardiometabolic disease (CMD), which encompasses metabolic-associated fatty liver disease (MAFLD), chronic kidney disease (CKD) and cardiovascular disease (CVD), has been increasing considerably in the past 50 years. CMD is a complex disease that can be influenced by genetics and environmental factors such as diet. With the increased reliance on processed foods containing saturated fats, fructose and cholesterol, a mechanistic understanding of how these molecules cause metabolic disease is required. A major pathway by which excessive nutrients contribute to CMD is through oxidative stress. In this review, we discuss how oxidative stress can drive CMD and the role of aberrant nutrient metabolism and genetic risk factors and how they potentially interact to promote progression of MAFLD, CVD and CKD. This review will focus on genetic mutations that are known to alter nutrient metabolism. We discuss the major genetic risk factors for MAFLD, which include Patatin-like phospholipase domain-containing protein 3 (PNPLA3), Membrane Bound O-Acyltransferase Domain Containing 7 (MBOAT7) and Transmembrane 6 Superfamily Member 2 (TM6SF2). In addition, mutations that prevent nutrient uptake cause hypercholesterolemia that contributes to CVD. We also discuss the mechanisms by which MAFLD, CKD and CVD are mutually associated with one another. In addition, some of the genetic risk factors which are associated with MAFLD and CVD are also associated with CKD, while some genetic risk factors seem to dissociate one disease from the other. Through a better understanding of the causative effect of genetic mutations in CMD and how aberrant nutrient metabolism intersects with our genetics, novel therapies and precision approaches can be developed for treating CMD.
Collapse
Affiliation(s)
- Andrew J. Butcko
- Hypertension and Vascular Research Division, Henry Ford Hospital, 6135 Woodward Avenue, Detroit, MI 48202, USA; (A.J.B.); (A.K.P.)
- Department of Physiology, Wayne State University, 540 E. Canfield Street, Detroit, MI 48202, USA
| | - Ashley K. Putman
- Hypertension and Vascular Research Division, Henry Ford Hospital, 6135 Woodward Avenue, Detroit, MI 48202, USA; (A.J.B.); (A.K.P.)
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, 784 Wilson Road, East Lansing, MI 48823, USA
| | - Emilio P. Mottillo
- Hypertension and Vascular Research Division, Henry Ford Hospital, 6135 Woodward Avenue, Detroit, MI 48202, USA; (A.J.B.); (A.K.P.)
- Department of Physiology, Wayne State University, 540 E. Canfield Street, Detroit, MI 48202, USA
| |
Collapse
|
15
|
Charchar FJ, Prestes PR, Mills C, Ching SM, Neupane D, Marques FZ, Sharman JE, Vogt L, Burrell LM, Korostovtseva L, Zec M, Patil M, Schultz MG, Wallen MP, Renna NF, Islam SMS, Hiremath S, Gyeltshen T, Chia YC, Gupta A, Schutte AE, Klein B, Borghi C, Browning CJ, Czesnikiewicz-Guzik M, Lee HY, Itoh H, Miura K, Brunström M, Campbell NR, Akinnibossun OA, Veerabhadrappa P, Wainford RD, Kruger R, Thomas SA, Komori T, Ralapanawa U, Cornelissen VA, Kapil V, Li Y, Zhang Y, Jafar TH, Khan N, Williams B, Stergiou G, Tomaszewski M. Lifestyle management of hypertension: International Society of Hypertension position paper endorsed by the World Hypertension League and European Society of Hypertension. J Hypertens 2024; 42:23-49. [PMID: 37712135 PMCID: PMC10713007 DOI: 10.1097/hjh.0000000000003563] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023]
Abstract
Hypertension, defined as persistently elevated systolic blood pressure (SBP) >140 mmHg and/or diastolic blood pressure (DBP) at least 90 mmHg (International Society of Hypertension guidelines), affects over 1.5 billion people worldwide. Hypertension is associated with increased risk of cardiovascular disease (CVD) events (e.g. coronary heart disease, heart failure and stroke) and death. An international panel of experts convened by the International Society of Hypertension College of Experts compiled lifestyle management recommendations as first-line strategy to prevent and control hypertension in adulthood. We also recommend that lifestyle changes be continued even when blood pressure-lowering medications are prescribed. Specific recommendations based on literature evidence are summarized with advice to start these measures early in life, including maintaining a healthy body weight, increased levels of different types of physical activity, healthy eating and drinking, avoidance and cessation of smoking and alcohol use, management of stress and sleep levels. We also discuss the relevance of specific approaches including consumption of sodium, potassium, sugar, fibre, coffee, tea, intermittent fasting as well as integrated strategies to implement these recommendations using, for example, behaviour change-related technologies and digital tools.
Collapse
Affiliation(s)
- Fadi J. Charchar
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
- Department of Physiology, University of Melbourne, Melbourne, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Priscilla R. Prestes
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Charlotte Mills
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Siew Mooi Ching
- Department of Family Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang
- Department of Medical Sciences, School of Medical and Live Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - Dinesh Neupane
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, USA
| | - Francine Z. Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne
| | - James E. Sharman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Liffert Vogt
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Louise M. Burrell
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Lyudmila Korostovtseva
- Department of Hypertension, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Manja Zec
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, USA
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Mansi Patil
- Department of Nutrition and Dietetics, Asha Kiran JHC Hospital, Chinchwad
- Hypertension and Nutrition, Core Group of IAPEN India, India
| | - Martin G. Schultz
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | | | - Nicolás F. Renna
- Unit of Hypertension, Hospital Español de Mendoza, School of Medicine, National University of Cuyo, IMBECU-CONICET, Mendoza, Argentina
| | | | - Swapnil Hiremath
- Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Tshewang Gyeltshen
- Graduate School of Public Health, St. Luke's International University, Tokyo, Japan
| | - Yook-Chin Chia
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Selangor
- Department of Primary Care Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Abhinav Gupta
- Department of Medicine, Acharya Shri Chander College of Medical Sciences and Hospital, Jammu, India
| | - Aletta E. Schutte
- School of Population Health, University of New South Wales, The George Institute for Global Health, Sydney, New South Wales, Australia
- Hypertension in Africa Research Team, SAMRC Unit for Hypertension and Cardiovascular Disease, North-West University
- SAMRC Developmental Pathways for Health Research Unit, School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Britt Klein
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Claudio Borghi
- Department of Medical and Surgical Sciences, Faculty of Medicine, University of Bologna, Bologna, Italy
| | - Colette J. Browning
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Marta Czesnikiewicz-Guzik
- School of Medicine, Dentistry and Nursing-Dental School, University of Glasgow, UK
- Department of Periodontology, Prophylaxis and Oral Medicine; Jagiellonian University, Krakow, Poland
| | - Hae-Young Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hiroshi Itoh
- Department of Internal Medicine (Nephrology, Endocrinology and Metabolism), Keio University, Tokyo
| | - Katsuyuki Miura
- NCD Epidemiology Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Mattias Brunström
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Norm R.C. Campbell
- Libin Cardiovascular Institute, Department of Medicine, University of Calgary, Calgary, Canada
| | | | - Praveen Veerabhadrappa
- Kinesiology, Division of Science, The Pennsylvania State University, Reading, Pennsylvania
| | - Richard D. Wainford
- Department of Pharmacology and Experimental Therapeutics, The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston
- Division of Cardiology, Emory University, Atlanta, USA
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Shane A. Thomas
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Takahiro Komori
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Udaya Ralapanawa
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | | | - Vikas Kapil
- William Harvey Research Institute, Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, BRC, Faculty of Medicine and Dentistry, Queen Mary University London
- Barts BP Centre of Excellence, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai
| | - Yuqing Zhang
- Department of Cardiology, Fu Wai Hospital, Chinese Academy of Medical Sciences, Chinese Hypertension League, Beijing, China
| | - Tazeen H. Jafar
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore
- Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| | - Nadia Khan
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Bryan Williams
- University College London (UCL), Institute of Cardiovascular Science, National Institute for Health Research (NIHR), UCL Hospitals Biomedical Research Centre, London, UK
| | - George Stergiou
- Hypertension Centre STRIDE-7, School of Medicine, Third Department of Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester
- Manchester Academic Health Science Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
16
|
Zhang R, Jadhav DA, Kramer B, Gonzalez-Vicente A. Profiling cellular heterogeneity and fructose transporter expression in the rat nephron by integrating single-cell and microdissected tubule segment transcriptomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572656. [PMID: 38187558 PMCID: PMC10769391 DOI: 10.1101/2023.12.20.572656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Single-cell RNA sequencing (scRNAseq) is a crucial tool in kidney research. These technologies cluster cells according to transcriptome similarity, irrespective of the anatomical location and ordering within the nephron. Thus, a cluster transcriptome may obscure heterogeneity of the cell population within a nephron segment. Elevated dietary fructose leads to salt-sensitive hypertension, in part by fructose reabsorption in the proximal tubule (PT). However, organization of the four known fructose transporters in apical PTs (SGLT4, SGLT5, GLUT5 and NaGLT1) remains poorly understood. We hypothesized that cells within each subsegment of the proximal tubule exhibit complex, heterogenous fructose transporter expression patterns. To test this hypothesis we analyzed rat and kidney transcriptomes and proteomes from publicly available scRNAseq and tubule microdissection databases. We found that microdissected PT-S1 segments consist of 81±12% cells with scRNAseq-derived transcriptional characteristics of S1, whereas PT-S2 express a mixture of 18±9% S1, 58±8% S2, and 19±5% S3 transcripts, and PT-S3 consists of 75±9% S3 transcripts. The expression of all four fructose transporters was detectable in all three PT segments, but key fructose transporters SGLT5 and GLUT5 progressively increased from S1 to S3, and both were significantly upregulated in S3 vs. S1/S2 (Slc5a10: 1.9 log 2 FC, p<1×10 -299 ; Scl2a5: 1.4 log 2 FC, p<4×10 -105 ). A similar distribution was found in human kidneys. These data suggest that S3 is the primary site of fructose reabsorption in both humans and rats. Finally, because of the multiple scRNAseq transcriptional phenotypes found in each segment our findings also imply that anatomic labels applied to scRNAseq clusters may be misleading.
Collapse
|
17
|
Li Y, Jiang T, Du M, He S, Huang N, Cheng B, Yan C, Tang W, Gao W, Guo H, Li Q, Wang Q. Ketohexokinase-dependent metabolism of cerebral endogenous fructose in microglia drives diabetes-associated cognitive dysfunction. Exp Mol Med 2023; 55:2417-2432. [PMID: 37907746 PMCID: PMC10689812 DOI: 10.1038/s12276-023-01112-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 11/02/2023] Open
Abstract
Dementia, as an advanced diabetes-associated cognitive dysfunction (DACD), has become the second leading cause of death among diabetes patients. Given that little guidance is currently available to address the DACD process, it is imperative to understand the underlying mechanisms and screen out specific therapeutic targets. The excessive endogenous fructose produced under high glucose conditions can lead to metabolic syndrome and peripheral organ damage. Although generated by the brain, the role of endogenous fructose in the exacerbation of cognitive dysfunction is still unclear. Here, we performed a comprehensive study on leptin receptor-deficient T2DM mice and their littermate m/m mice and revealed that 24-week-old db/db mice had cognitive dysfunction and excessive endogenous fructose metabolism in the hippocampus by multiomics analysis and further experimental validation. We found that the rate-limiting enzyme of fructose metabolism, ketohexokinase, is primarily localized in microglia. It is upregulated in the hippocampus of db/db mice, which enhances mitochondrial damage and reactive oxygen species production by promoting nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) expression and mitochondrial translocation. Inhibiting fructose metabolism via ketohexokinase depletion reduces microglial activation, leading to the restoration of mitochondrial homeostasis, recovery of structural synaptic plasticity, improvement of CA1 pyramidal neuron electrophysiology and alleviation of cognitive dysfunction. Our findings demonstrated that enhanced endogenous fructose metabolism in microglia plays a dominant role in diabetes-associated cognitive dysfunction and could become a potential target for DACD.
Collapse
Affiliation(s)
- Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Tao Jiang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, Shaanxi, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Shuxuan He
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Ning Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Wenxin Tang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Hongyan Guo
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Qiao Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
18
|
Mao D, Feng J, Zhou Y, Li H. Analysis of different plant- and animal-based dietary patterns and their relationship with serum uric acid levels in Chinese adults. Nutr J 2023; 22:53. [PMID: 37891672 PMCID: PMC10605343 DOI: 10.1186/s12937-023-00885-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Dietary patterns play an important role in regulating serum uric acid levels in the body, but evidence for the association between different kinds of plant-based and animal-based dietary patterns and individual serum uric acid levels is scarce and inconsistent. METHODS We analyzed data from the sixth wave of the China Health and Nutrition Survey. The plant-based diet of 7,806 participants was determined using three consecutive 24-hour dietary recalls, and latent profile analysis was used to identify dietary patterns among participants. Serum uric acid levels were analyzed using the enzymatic colorimetric method. The association between intakes of different types of dietary pattern and individual serum uric acid levels was analyzed using linear regression analysis, after adjusting for confounding variables. RESULTS We identified three types of plant-based dietary patterns, namely, low tuber starches and vegetable plant-based diet (LTVP), high cereal, tuber starches and vegetable plant-based diet (HCTVP), and high legume and fruit plant-based diet (HLFP). We also identified three types of animal-based dietary patterns, namely, high milk and egg animal-based diet (HMiEA), low egg and fish animal-based diet, and high meat and fish animal-based diet (HMeFA). Significant coefficients for participant serum uric acid levels were observed for the HCTVP diet (β = -0.022, P = 0.031) and HMeFA diet (β = 0.061, P < 0.001). The median intake of foods in the HCTVP diet was as follows: cereals and cereal products, 444.83 g/d; tubers and starch products, 166.67 g/d; dried legumes and legume products, 8.33 g/d; vegetables and vegetable products, 333.33 g/d; and fruits and fruit products, 0 g/d. The median intake of foods in the HMeFA diet was as follows: meat and meat products, 73.33 g/d; poultry and poultry products, 0 g/d; milk and milk products, 0 g/d; eggs and egg products, 26.67 g/d; and fish, shellfish, and mollusks, 180.00 g/d. CONCLUSION We showed that individual serum uric acid levels (1) might decrease under the plant-based HCTVP diet, (2) might increase under the animal-based HMeFA diet, (3) might not decrease under the plant-based HLFP diet, and (4) might not increase under the animal-based HMiEA diet. Further studies are needed to confirm these associations.
Collapse
Affiliation(s)
- Danhui Mao
- Health Management and Policy Research Center, School of Management, Shanxi Medical University, 030000, Taiyuan, China.
| | - Jin Feng
- Health Management and Policy Research Center, School of Management, Shanxi Medical University, 030000, Taiyuan, China
| | - Yangzilin Zhou
- Health Management and Policy Research Center, School of Management, Shanxi Medical University, 030000, Taiyuan, China
| | - Honggang Li
- Health Management and Policy Research Center, School of Management, Shanxi Medical University, 030000, Taiyuan, China
| |
Collapse
|
19
|
Romualdo GR, de Souza JLH, Valente LC, Barbisan LF. Assessment of the impact of glyphosate and 2,4-D herbicides on the kidney injury and transcriptome changes in obese mice fed a Western diet. Toxicol Lett 2023; 385:1-11. [PMID: 37567420 DOI: 10.1016/j.toxlet.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/21/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
The development of chronic kidney disease has been associated with comorbidities resulting from the consumption of Westernized dietary (WD) patterns, including obesity and other metabolic dysfunctions. Kidneys also have a crucial role in the metabolism and excretion of xenobiotics, including herbicides. There is limited knowledge regarding the simultaneous exposure to WD and glyphosate (glypho) and 2,4-D, the most used herbicides globally. Thus, this study examined whether exposure to glypho and/or 2,4-D, either individually or in mixed, could impact the early effects of WD intake on kidney histology and gene expression in a rodent model. Male C57BL6J mice were fed a WD containing 20% lard, 0.2% cholesterol, 20% sucrose, and high sugar solution with 23.1 and 18.9 g/L of D-fructose and D-glucose for six months. During this period, the mice also received glypho (0.05 or 5 mg/kg/day), 2,4-D (0.02 or 2 mg/kg/day), or a mixture of both (0.05 +0.02, 5 +2 mg/kg/day) via intragastric administration five times per week. The doses were within or below the established regulatory limits. While single or mixed exposures did not alter WD-induced obesity, tubular lipid vacuolation, or increased serum creatinine levels; the exposure to higher doses of the mixture (5 +2) reduced the mesangial matrix area and tubular cell proliferation, while increasing the density of F4/80 macrophages in the renal interstitium. In terms of transcriptomic analysis, the herbicide mixture altered the expression of 415 genes in the kidney, which were found to be associated with immune response processes, particularly those related to phagocyte activity. While discrete, findings indicate that herbicide mixtures, rather than single exposures, might induce minor deleterious effects on the kidneys of obese mice under WD intake.
Collapse
Affiliation(s)
- Guilherme Ribeiro Romualdo
- São Paulo State University (UNESP), Botucatu Medical School, Experimental Research Unit (UNIPEX), Multimodel Drug Screening Platform - Laboratory of Chemically Induced and Experimental Carcinogenesis (MDSP-LCQE), Botucatu, SP, Brazil; São Paulo State University (UNESP), Biosciences Institute, Department of Structural and Functional Biology, Laboratory of Chemically Induced and Experimental Carcinogenesis (LCQE), Botucatu, SP, Brazil
| | - Jéssica Luri Hisano de Souza
- São Paulo State University (UNESP), Biosciences Institute, Department of Structural and Functional Biology, Laboratory of Chemically Induced and Experimental Carcinogenesis (LCQE), Botucatu, SP, Brazil
| | - Letícia Cardoso Valente
- São Paulo State University (UNESP), Botucatu Medical School, Experimental Research Unit (UNIPEX), Multimodel Drug Screening Platform - Laboratory of Chemically Induced and Experimental Carcinogenesis (MDSP-LCQE), Botucatu, SP, Brazil; São Paulo State University (UNESP), Biosciences Institute, Department of Structural and Functional Biology, Laboratory of Chemically Induced and Experimental Carcinogenesis (LCQE), Botucatu, SP, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Botucatu, SP, Brazil
| | - Luís Fernando Barbisan
- São Paulo State University (UNESP), Biosciences Institute, Department of Structural and Functional Biology, Laboratory of Chemically Induced and Experimental Carcinogenesis (LCQE), Botucatu, SP, Brazil.
| |
Collapse
|
20
|
Estevez H, Garcia-Calvo E, Mena ML, Alvarez-Fernandez Garcia R, Luque-Garcia JL. Unraveling the Mechanisms of Ch-SeNP Cytotoxicity against Cancer Cells: Insights from Targeted and Untargeted Metabolomics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2204. [PMID: 37570523 PMCID: PMC10420838 DOI: 10.3390/nano13152204] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
Abstract
Although chitosan-stabilized selenium nanoparticles (Ch-SeNPs) have emerged as a promising chemical form of selenium for anticancer purposes, gathering more profound knowledge related to molecular dysfunctions contributes significantly to the promotion of their evolution as a chemotherapeutic drug. In this sense, metabolites are the end products in the flow of gene expression and, thus, the most sensitive to changes in the physiological state of a biological system. Therefore, metabolomics provides a functional readout of the biochemical activity and cell state. In the present study, we evaluated alterations in the metabolomes of HepG2 cells after the exposure to Ch-SeNPs to elucidate the biomolecular mechanisms involved in their therapeutic effect. A targeted metabolomic approach was conducted to evaluate the levels of four of the main energy-related metabolites (adenosine triphosphate (ATP); adenosine diphosphate (ADP); nicotinamide adenine dinucleotide (NAD+); and 1,4-dihydronicotinamide adenine dinucleotide (NADH)), revealing alterations as a result of exposure to Ch-SeNPs related to a shortage in the energy supply system in the cell. In addition, an untargeted metabolomic experiment was performed, which allowed for the study of alterations in the global metabolic profile as a consequence of Ch-SeNP exposure. The results indicate that the TCA cycle and glycolytic pathways were impaired, while alternative pathways such as glutaminolysis and cysteine metabolism were upregulated. Additionally, increased fructose levels suggested the induction of hypoxia-like conditions. These findings highlight the potential of Ch-SeNPs to disrupt cancer cell metabolism and provide insights into the mechanisms underlying their antitumor effects.
Collapse
Affiliation(s)
| | | | | | | | - Jose L. Luque-Garcia
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (H.E.); (E.G.-C.); (M.L.M.); (R.A.-F.G.)
| |
Collapse
|
21
|
Nysather J, Kaya E, Manka P, Gudsoorkar P, Syn WK. Nonalcoholic Fatty Liver Disease and Chronic Kidney Disease Cross Talk. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:315-335. [PMID: 37657879 DOI: 10.1053/j.akdh.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/14/2022] [Accepted: 04/04/2023] [Indexed: 09/03/2023]
Abstract
Nonalcoholic fatty liver disease is a multisystem condition with effects beyond the liver. The identification of chronic kidney disease as an independent mediator of nonalcoholic fatty liver disease or associated entity with shared cardiometabolic risk factors remains controversial and continues to draw scientific interest. With increasing prevalence of nonalcoholic fatty liver disease and lack of Food and Drug Administration approved therapies, these shared cardiometabolic risk factors have drawn significant attention. In this article, we review shared pathophysiological mechanisms between nonalcoholic fatty liver disease and chronic kidney disease along with current treatment strategies that might be useful for both disease processes.
Collapse
Affiliation(s)
- Jacob Nysather
- Division of Nephrology and Kidney C.A.R.E. Program, University of Cincinnati, OH
| | - Eda Kaya
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Paul Manka
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Prakash Gudsoorkar
- Division of Nephrology and Kidney C.A.R.E. Program, University of Cincinnati, OH
| | - Wing-Kin Syn
- Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO; Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Euskal Herriko Unibertsitatea/Universidad del País Vasco, Leioa, Spain.
| |
Collapse
|
22
|
Liu J, Pan H, Liu Y, Guan M, Li X, Chen S, Tong X, Luo Y, Wang X, Yang X, Guo X, Zhang J, Tao L. Distinct hyperuricemia trajectories are associated with different risks of incident diabetes: A prospective cohort study. Nutr Metab Cardiovasc Dis 2023; 33:967-977. [PMID: 36958974 DOI: 10.1016/j.numecd.2023.02.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND AND AIM Conflicting results suggest a link between serum uric acid and diabetes and previous studies ignored the effect of continuous exposure of serum uric acid on diabetes risk. This study aims to characterize hyperuricemia trajectories in middle-aged adults and to examine its potential impact on diabetes risk, considering the role of obesity, dyslipidemia, and hypertension. METHODS AND RESULTS The cohort included 9192 participants who were free of diabetes before 2013. The hyperuricemia trajectories during 2009-2013 were identified by latent class growth models. Incident diabetes during 2014-2018 was used as the outcome. Modified Poisson regression models were used to assess the association of trajectories with diabetes. Furthermore, marginal structural models were used to estimate the mediating effects of the relationship between hyperuricemia trajectories and diabetes. We identified three discrete hyperuricemia trajectories: high-increasing (n = 5794), moderate-stable (n = 2049), and low-stable (n = 1349). During 5 years of follow-up, we documented 379 incident diabetes cases. Compared with the low-stable pattern, the high-increasing pattern had a higher risk of developing diabetes (RR, 1.42; 95% CI: 1.09-1.84). In addition, the percentages of total effect between the high-increasing hyperuricemia pattern and diabetes mediated by obesity, dyslipidemia, and hypertension were 24.41%, 18.26%, and 6.29%. However, the moderate-stable pattern was not associated with an increased risk of diabetes. CONCLUSIONS These results indicate that the high-increasing hyperuricemia trajectory is significantly associated with an increased risk of diabetes. Furthermore, obesity, dyslipidemia, and hypertension play mediating roles in the relationship between the high-increasing hyperuricemia pattern and increased diabetes risk.
Collapse
Affiliation(s)
- Jia Liu
- Yanjing Medical College, Capital Medical University, Beijing 101300, China
| | - Huiying Pan
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China; Centre for Precision Health, Edith Cowan University, Perth, WA 6027, Australia
| | - Yue Liu
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Mengying Guan
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne 3086, Australia
| | - Shuo Chen
- Department of Information, Beijing Physical Examination Center, Beijing 100077, China
| | - Xingyao Tong
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Yanxia Luo
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Xiaonan Wang
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Xinghua Yang
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Xiuhua Guo
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China; Centre for Precision Health, Edith Cowan University, Perth, WA 6027, Australia
| | - Jingbo Zhang
- Department of Information, Beijing Physical Examination Center, Beijing 100077, China.
| | - Lixin Tao
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
23
|
Shen L, Yang Y, Zhang J, Feng L, Zhou Q. Diacylated anthocyanins from purple sweet potato ( Ipomoeabatatas L.) attenuate hyperglycemia and hyperuricemia in mice induced by a high-fructose/high-fat diet. J Zhejiang Univ Sci B 2023; 24:587-601. [PMID: 37455136 PMCID: PMC10350372 DOI: 10.1631/jzus.b2200587] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/06/2023] [Indexed: 04/15/2023]
Abstract
Studies have shown that targeting xanthine oxidase (XO) can be a feasible treatment for fructose-induced hyperuricemia and hyperglycemia. This study aimed to evaluate the dual regulatory effects and molecular mechanisms of diacylated anthocyanins from purple sweet potato (diacylated AF-PSPs) on hyperglycemia and hyperuricemia induced by a high-fructose/high-fat diet. The body weight, organ index, serum biochemical indexes, and liver antioxidant indexes of mice were measured, and the kidneys were observed in pathological sections. The relative expression levels of messenger RNAs (mRNAs) of fructose metabolism pathway enzymes in kidney were detected by fluorescent real-time quantitative polymerase chain (qPCR) reaction technique, and the expression of renal transporter protein and inflammatory factor pathway protein was determined by immunohistochemistry (IHC) technique. Results showed that diacylated AF-PSPs alleviated hyperuricemia in mice, and that this effect might be related to the regulation of liver XO activity, lipid accumulation, and relevant renal transporters. Diacylated AF-PSPs reduced body weight and relieved lipid metabolism disorder, liver lipid accumulation, and liver oxidative stress, thereby enhancing insulin utilization and sensitivity, lowering blood sugar, and reducing hyperglycemia in mice. Also, diacylated AF-PSPs restored mRNA levels related to renal fructose metabolism, and reduced kidney injury and inflammation. This study provided experimental evidence for the mechanisms of dual regulation of blood glucose and uric acid (UA) by diacylated AF-PSPs and their utilization as functional foods in the management of metabolic syndrome.
Collapse
Affiliation(s)
- Luhong Shen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Yang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiuliang Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Lanjie Feng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Qing Zhou
- Department of Pharmacy, Wuhan City Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China.
| |
Collapse
|
24
|
Iizuka K. Recent Progress on Fructose Metabolism-Chrebp, Fructolysis, and Polyol Pathway. Nutrients 2023; 15:nu15071778. [PMID: 37049617 PMCID: PMC10096667 DOI: 10.3390/nu15071778] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Excess fructose intake is associated with obesity, fatty liver, tooth decay, cancer, and cardiovascular diseases. Even after the ingestion of fructose, fructose concentration in the portal blood is never high; fructose is further metabolized in the liver, and the blood fructose concentration is 1/100th of the glucose concentration. It was previously thought that fructose was metabolized in the liver and not in the small intestine, but it has been reported that metabolism in the small intestine also plays an important role in fructose metabolism. Glut5 knockout mice exhibit poor fructose absorption. In addition, endogenous fructose production via the polyol pathway has also received attention; gene deletion of aldose reductase (Ar), ketohexokinase (Khk), and triokinase (Tkfc) has been found to prevent the development of fructose-induced liver lipidosis. Carbohydrate response element-binding protein (Chrebp) regulates the expression of Glut5, Khk, aldolase b, and Tkfc. We review fructose metabolism with a focus on the roles of the glucose-activating transcription factor Chrebp, fructolysis, and the polyol pathway.
Collapse
Affiliation(s)
- Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan
- Food and Nutrition Service Department, Fujita Health University Hospital, Toyoake 470-1192, Japan
| |
Collapse
|
25
|
Sukiasyan L. Fructose-Induced Alteration of the Heart and Vessels Homeostasis. Curr Probl Cardiol 2023; 48:101013. [PMID: 34637847 DOI: 10.1016/j.cpcardiol.2021.101013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 01/04/2023]
Abstract
To date, the role of uncontrolled sugar consumption in the triggering and progression of cardiovascular events is undeniable. Modern concepts offer a new hypothesis regarding the direct myocardiotoxic effects of fructose. Experimental studies have demonstrated that cardiomyocytes have a unique ability to transport and use fructose along with the expression of all components involved in fructose metabolism. The purpose of this review article is to assess and analyze the available knowledge on fructose-induced cardiotoxicity detection since understanding the pathophysiological mechanisms and pathobiochemical aspects will become the basis for the determination of a rational myocardioprotection regimen.
Collapse
Affiliation(s)
- Lilit Sukiasyan
- Yerevan State Medical University after M.Heratsi, Armenia; L. A. Orbeli Institute of Human Physiology, Armenia.
| |
Collapse
|
26
|
Huynh C, Ryu J, Lee J, Inoki A, Inoki K. Nutrient-sensing mTORC1 and AMPK pathways in chronic kidney diseases. Nat Rev Nephrol 2023; 19:102-122. [PMID: 36434160 DOI: 10.1038/s41581-022-00648-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Nutrients such as glucose, amino acids and lipids are fundamental sources for the maintenance of essential cellular processes and homeostasis in all organisms. The nutrient-sensing kinases mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) are expressed in many cell types and have key roles in the control of cell growth, proliferation, differentiation, metabolism and survival, ultimately contributing to the physiological development and functions of various organs, including the kidney. Dysregulation of these kinases leads to many human health problems, including cancer, neurodegenerative diseases, metabolic disorders and kidney diseases. In the kidney, physiological levels of mTOR and AMPK activity are required to support kidney cell growth and differentiation and to maintain kidney cell integrity and normal nephron function, including transport of electrolytes, water and glucose. mTOR forms two functional multi-protein kinase complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Hyperactivation of mTORC1 leads to podocyte and tubular cell dysfunction and vulnerability to injury, thereby contributing to the development of chronic kidney diseases, including diabetic kidney disease, obesity-related kidney disease and polycystic kidney disease. Emerging evidence suggests that targeting mTOR and/or AMPK could be an effective therapeutic approach to controlling or preventing these diseases.
Collapse
Affiliation(s)
- Christopher Huynh
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jaewhee Ryu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jooho Lee
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ayaka Inoki
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA.,Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA. .,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Muacevic A, Adler JR. A Narrative Review of New Treatment Options for Diabetic Nephropathy. Cureus 2023; 15:e33235. [PMID: 36733548 PMCID: PMC9889842 DOI: 10.7759/cureus.33235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023] Open
Abstract
Diabetic nephropathy (DN) is a type of nephropathy that is caused by a diabetic condition. Diabetic nephropathy is seen in type 1 and type 2 diabetes. End-stage renal disorders are brought on by DN. Diabetic nephropathy is thought to be linked to metabolic changes in the body. Proteinuria and glomerular filtration rate are the two most crucial diagnostic and prognosis measures for diabetic kidney disease (DKD), yet both have significant disadvantages. Novel biomarkers are thus increasingly required to improve risk factors and detect disease at an early stage. Controlling blood glucose and vital sign like body temperature and blood pressure, reducing cholesterol levels, and blocking the renin-angiotensin system are the standard treatments for diabetic patients. On the other hand, if used too late within the course of the disease, these therapeutic techniques can only provide partial relief from nephropathy. The complicated pathophysiology of the diabetic kidney, which experiences a variety of severe structural, metabolic, and functional alterations, represents one of the most important obstacles to the event of effective therapeutics for DN. Despite these issues, new diabetes models have identified promising treatment targets by identifying the mechanisms that control important functions of podocytes and glomerular endothelial cells. It has been shown in the vast majority of trials that renin-angiotensin system inhibitors combined with integrative therapies work well for DN. Combining sodium-glucose cotransporter-2 inhibitors and renin-angiotensin-aldosterone system blockers is a novel way to slow down the course of DKD by lowering inflammatory and fibrotic indicators brought on by hyperglycemia, which is more effective than using either medicine alone. Aldosterone receptor inhibitors and advanced glycation end-product inhibitors are two recently produced medications that may be used successfully to treat DN.
Collapse
|
28
|
Courville K, Bustamante N, Hurtado B, Pecchio M, Rodríguez C, Núñez-Samudio V, Landires I. Chronic kidney disease of nontraditional causes in central Panama. BMC Nephrol 2022; 23:275. [PMID: 35931963 PMCID: PMC9356394 DOI: 10.1186/s12882-022-02907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 08/03/2022] [Indexed: 11/24/2022] Open
Abstract
Background Over the last three decades, the mesoamerican region has seen an increase in the frequency of patients diagnosed with Chronic Kidney Disease of nontraditional causes (CKDnt) also known as Meso-American Nephropathy (MeN). A region with an increased frequency of patients with Chronic Kidney Disease (CKD) has been identified in central Panama. The present study aims to characterize the clinical presentation of patients with CKDnt in an understudied population of the central region of Panama and to compare them with patients with traditional chronic kidney disease (CKDt). Methods A retrospective descriptive study was conducted in a nephrology reference hospital in the central provinces of Herrera and Los Santos, comparing a group of 15 patients with CKDnt to 91 patients with CKDt. Sociodemographic variables, personal history, laboratory parameters, and of renal ultrasound were compared. Results Patients with CKDnt had a median age of 58 years (IQR: 52–61), significantly lower (P < 0.001) than patients with CKDt with a median age of 71 years (IQR: 64–78). Patients with CKDnt had a history of being agricultural (60%) and transportation (20%) workers, significantly higher than patients with CKDt (15%, P < 0.001 and 0%, P < 0.01 respectively). Renal atrophy and hyperuricemia are significant clinical markers of CKDnt (P < 0.001 and P < 0.05 respectively). Conclusion To our knowledge, this is the first study in Panama to investigate the clinical presentation of patients with CKDnt and one of the few in Central America and the world that compares them with patients with CKDt. In central Panama the typical CKDnt patient is a male in his 50 s who is primarily engaged in agriculture or as a public transport driver. Renal atrophy and hyperuricemia are significant clinical markers of CKDnt. Further studies are needed to help understand the common determinants and risk factors for CKDnt development in Panama and Mesoamerica.
Collapse
Affiliation(s)
- Karen Courville
- Instituto de Ciencias Médicas, PO Box 0710-00043, Las Tablas, Los Santos, Panamá.,Unidad de Hemodiálisis, Departamento de Nefrología, Hospital Dr. Gustavo N. Collado, Caja de Seguro Social, Chitré, Herrera, Panamá
| | - Norman Bustamante
- Instituto de Ciencias Médicas, PO Box 0710-00043, Las Tablas, Los Santos, Panamá.,Unidad de Hemodiálisis, Departamento de Nefrología, Hospital Dr. Gustavo N. Collado, Caja de Seguro Social, Chitré, Herrera, Panamá
| | - Bárbara Hurtado
- Departamento de Epidemiología, Hospital Dr. Gustavo N. Collado, Caja de Seguro Social, Chitré, Herrera, Panamá
| | - Maydelin Pecchio
- Instituto de Ciencias Médicas, PO Box 0710-00043, Las Tablas, Los Santos, Panamá.,Unidad de Infectología, Hospital Dr. Gustavo Nelson Collado, Caja de Seguro Social, Chitré, Herrera, Panamá
| | - Clarissa Rodríguez
- Departamento de Infecciones Nosocomiales, Hospital Dr. Gustavo N. Collado, Caja de Seguro Social, Chitré, Herrera, Panamá
| | - Virginia Núñez-Samudio
- Instituto de Ciencias Médicas, PO Box 0710-00043, Las Tablas, Los Santos, Panamá. .,Sección de Epidemiología, Departamento de Salud Pública, Región de Salud de Herrera, Ministerio de Salud, Chitré, Herrera, Panamá.
| | - Iván Landires
- Instituto de Ciencias Médicas, PO Box 0710-00043, Las Tablas, Los Santos, Panamá. .,Hospital Joaquín Pablo Franco Sayas, Ministerio de Salud, Las Tablas, Los Santos, Panamá.
| |
Collapse
|
29
|
Ultrasound Assessment of the Relevance of Liver, Spleen, and Kidney Dimensions with Body Parameters in Adolescents. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9150803. [PMID: 35832132 PMCID: PMC9273413 DOI: 10.1155/2022/9150803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022]
Abstract
Objective Ultrasound is a practical imaging modality for screening and identification of anomalies in the organs. This study used ultrasonography to examine the association between body parameters and dimensions of the normal liver, spleen, and kidney in adolescents based on ultrasound examination results. Methods A total of 300 junior and senior high school teenagers receiving routine health check-ups in our hospital from January 2020 to January 2021 were included. Their height and weight were measured, and their body surface area (BSA) and body mass index (BMI) were calculated. Ultrasound imaging was employed to obtain information such as the length and volume of the liver, gallbladder, spleen, and kidney. Besides, the correlation of body parameters such as gender, age, height, weight, BSA, and BMI with visceral dimension was investigated using the Pearson test and multiple regression analysis, respectively. Results We observed that the abdominal organs of adolescents were enlarged with age. The span and volume of the liver and the length and volume of the right kidney were significantly larger in boys than in girls. The age, BSA, and BMI were positively correlated with the liver span and spleen length, as well as the left and right kidney lengths. Additionally, age, BSA, and BMI were identified as important predictors for dimensions of the spleen, liver, and kidney. Conclusions Body parameters are notably associated with the dimensions of the liver, spleen, and kidney and could be utilized as predicting factors for the liver, spleen, and kidney dimensions.
Collapse
|
30
|
Fang XY, Qi LW, Chen HF, Gao P, Zhang Q, Leng RX, Fan YG, Li BZ, Pan HF, Ye DQ. The Interaction Between Dietary Fructose and Gut Microbiota in Hyperuricemia and Gout. Front Nutr 2022; 9:890730. [PMID: 35811965 PMCID: PMC9257186 DOI: 10.3389/fnut.2022.890730] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
With the worldwide epidemics of hyperuricemia and associated gout, the diseases with purine metabolic disorders have become a serious threat to human public health. Accumulating evidence has shown that they have been linked to increased consumption of fructose in humans, we hereby made a timely review on the roles of fructose intake and the gut microbiota in regulating purine metabolism, together with the potential mechanisms by which excessive fructose intake contributes to hyperuricemia and gout. To this end, we focus on the understanding of the interaction between a fructose-rich diet and the gut microbiota in hyperuricemia and gout to seek for safe, cheap, and side-effect-free clinical interventions. Furthermore, fructose intake recommendations for hyperuricemia and gout patients, as well as the variety of probiotics and prebiotics with uric acid-lowering effects targeting the intestinal tract are also summarized to provide reference and guidance for the further research.
Collapse
Affiliation(s)
- Xin-yu Fang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Liang-wei Qi
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Hai-feng Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Peng Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Qin Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Rui-xue Leng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Yin-guang Fan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Bao-zhu Li
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Hai-feng Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Dong-qing Ye
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
- *Correspondence: Dong-qing Ye
| |
Collapse
|
31
|
Hansen J, Sealfon R, Menon R, Eadon MT, Lake BB, Steck B, Anjani K, Parikh S, Sigdel TK, Zhang G, Velickovic D, Barwinska D, Alexandrov T, Dobi D, Rashmi P, Otto EA, Rivera M, Rose MP, Anderton CR, Shapiro JP, Pamreddy A, Winfree S, Xiong Y, He Y, de Boer IH, Hodgin JB, Barisoni L, Naik AS, Sharma K, Sarwal MM, Zhang K, Himmelfarb J, Rovin B, El-Achkar TM, Laszik Z, He JC, Dagher PC, Valerius MT, Jain S, Satlin LM, Troyanskaya OG, Kretzler M, Iyengar R, Azeloglu EU. A reference tissue atlas for the human kidney. SCIENCE ADVANCES 2022; 8:eabn4965. [PMID: 35675394 PMCID: PMC9176741 DOI: 10.1126/sciadv.abn4965] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/20/2022] [Indexed: 05/08/2023]
Abstract
Kidney Precision Medicine Project (KPMP) is building a spatially specified human kidney tissue atlas in health and disease with single-cell resolution. Here, we describe the construction of an integrated reference map of cells, pathways, and genes using unaffected regions of nephrectomy tissues and undiseased human biopsies from 56 adult subjects. We use single-cell/nucleus transcriptomics, subsegmental laser microdissection transcriptomics and proteomics, near-single-cell proteomics, 3D and CODEX imaging, and spatial metabolomics to hierarchically identify genes, pathways, and cells. Integrated data from these different technologies coherently identify cell types/subtypes within different nephron segments and the interstitium. These profiles describe cell-level functional organization of the kidney following its physiological functions and link cell subtypes to genes, proteins, metabolites, and pathways. They further show that messenger RNA levels along the nephron are congruent with the subsegmental physiological activity. This reference atlas provides a framework for the classification of kidney disease when multiple molecular mechanisms underlie convergent clinical phenotypes.
Collapse
Affiliation(s)
- Jens Hansen
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Sealfon
- Princeton University, Princeton, NJ, USA
- Flatiron Institute, New York, NY, USA
| | - Rajasree Menon
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | | | - Blue B. Lake
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Becky Steck
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kavya Anjani
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Samir Parikh
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Tara K. Sigdel
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Guanshi Zhang
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
| | | | - Daria Barwinska
- Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Dejan Dobi
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Priyanka Rashmi
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Edgar A. Otto
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Miguel Rivera
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Michael P. Rose
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Christopher R. Anderton
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - John P. Shapiro
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Annapurna Pamreddy
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
| | - Seth Winfree
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuguang Xiong
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yongqun He
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Ian H. de Boer
- Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
| | | | | | - Abhijit S. Naik
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kumar Sharma
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
| | - Minnie M. Sarwal
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Jonathan Himmelfarb
- Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
| | - Brad Rovin
- Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Zoltan Laszik
- University of California San Francisco School of Medicine, San Francisco, CA, USA
| | | | | | - M. Todd Valerius
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Sanjay Jain
- Washington University in Saint Louis School of Medicine, St. Louis, MS, USA
| | - Lisa M. Satlin
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olga G. Troyanskaya
- Princeton University, Princeton, NJ, USA
- Flatiron Institute, New York, NY, USA
| | | | - Ravi Iyengar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Kidney Precision Medicine Project
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Princeton University, Princeton, NJ, USA
- Flatiron Institute, New York, NY, USA
- University of Michigan School of Medicine, Ann Arbor, MI, USA
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- University of California San Francisco School of Medicine, San Francisco, CA, USA
- Ohio State University College of Medicine, Columbus, OH, USA
- University of Texas–Health San Antonio School of Medicine, San Antonio, TX, USA
- Pacific Northwest National Laboratory, Richland, WA, USA
- European Molecular Biology Laboratory, Heidelberg, Germany
- Schools of Medicine and Public Health, University of Washington, Seattle, WA, USA
- Duke University School of Medicine, Durham, NC, USA
- Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- Washington University in Saint Louis School of Medicine, St. Louis, MS, USA
| |
Collapse
|
32
|
Hamada S, Takata T, Yamada K, Yamamoto M, Mae Y, Iyama T, Ikeda S, Kanda T, Sugihara T, Isomoto H. Steatosis is involved in the progression of kidney disease in a high-fat-diet-induced non-alcoholic steatohepatitis mouse model. PLoS One 2022; 17:e0265461. [PMID: 35294499 PMCID: PMC8926260 DOI: 10.1371/journal.pone.0265461] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
Chronic kidney disease (CKD) and non-alcoholic steatohepatitis (NASH) are major health issues associated with the metabolic syndrome. Although NASH is a known risk factor of CKD, the mechanisms linking these two diseases remain poorly understood. We aimed to investigate alterations in the kidney complicated with dyslipidemia in an established NASH mouse model. Male C57BL6/J mice were fed with control diet or high-fat diet (HFD), containing 40% fat, 22% fructose, and 2% cholesterol for 16 weeks. Metabolic characteristics, histological changes in the kidney, endoplasmic reticulum (ER) stress, apoptosis, and fibrosis were evaluated by histological analysis, immunoblotting, and quantitative reverse transcription-polymerase chain reaction. Levels of serum aspartate aminotransferase, alanine aminotransferase, alkali-phosphatase, total cholesterol, and urinary albumin were significantly higher in mice fed with HFD. Remarkable steatosis, glomerular hypertrophy, and interstitial fibrosis were also shown in in the kidney by leveraging HFD. Furthermore, HFD increased the mRNA expression levels of Casp3, Tgfb1, and Nfe2l2 and the protein level of BiP. We observed the early changes of CKD and speculate that the underlying mechanisms that link CKD and NASH are the induction of ER stress and apoptosis. Further, we observed the activation of Nfe2l2 in the steatosis-induced CKD mouse model. This NASH model holds implications in investigating the mechanisms linking dyslipidemia and CKD.
Collapse
Affiliation(s)
- Shintaro Hamada
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomoaki Takata
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
- * E-mail:
| | - Kentaro Yamada
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Marie Yamamoto
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yukari Mae
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Takuji Iyama
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Suguru Ikeda
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tsutomu Kanda
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Takaaki Sugihara
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
33
|
Rodriguez-Iturbe B, Johnson RJ, Lanaspa MA, Nakagawa T, Garcia-Arroyo FE, Sánchez-Lozada LG. Sirtuin deficiency and the adverse effects of fructose and uric acid synthesis. Am J Physiol Regul Integr Comp Physiol 2022; 322:R347-R359. [PMID: 35271385 PMCID: PMC8993531 DOI: 10.1152/ajpregu.00238.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 12/17/2022]
Abstract
Fructose metabolism and hyperuricemia have been shown to drive insulin resistance, metabolic syndrome, hepatic steatosis, hypertension, inflammation, and innate immune reactivity in experimental studies. We suggest that these adverse effects are at least in part the result of suppressed activity of sirtuins, particularly Sirtuin1. Deficiency of sirtuin deacetylations is a consequence of reduced bioavailability of its cofactor nicotinamide adenine dinucleotide (NAD+). Uric acid-induced inflammation and oxidative stress consume NAD+ and activation of the polyol pathway of fructose and uric acid synthesis also reduces the NAD+-to-NADH ratio. Variability in the compensatory regeneration of NAD+ could result in variable recovery of sirtuin activity that may explain the inconsistent benefits of treatments directed to reduce uric acid in clinical trials. Here, we review the pathogenesis of the metabolic dysregulation driven by hyperuricemia and their potential relationship with sirtuin deficiency. In addition, we discuss therapeutic options directed to increase NAD+ and sirtuins activity that may improve the adverse effects resulting from fructose and uric acid synthesis.
Collapse
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán," Mexico City, Mexico
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Denver, Colorado
- Kidney Disease Division, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, Colorado
| | - Miguel A Lanaspa
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon
| | | | - Fernando E Garcia-Arroyo
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| | - Laura G Sánchez-Lozada
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| |
Collapse
|
34
|
Bier A, Shapira E, Khasbab R, Sharabi Y, Grossman E, Leibowitz A. High-Fructose Diet Increases Renal ChREBPβ Expression, Leading to Intrarenal Fat Accumulation in a Rat Model with Metabolic Syndrome. BIOLOGY 2022; 11:biology11040618. [PMID: 35453816 PMCID: PMC9027247 DOI: 10.3390/biology11040618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 01/07/2023]
Abstract
Simple Summary Fructose consumption leads to the development of metabolic syndrome. Fatty liver and chronic kidney disease are closely related to metabolic syndrome. Lately, a transcription factor that regulates fructose metabolism in the liver, named ChREBPβ, which is responsible for de-novo lipogenesis and intra-hepatic fat accumulation (“fatty liver”), was described. In this study, we demonstrate that the effect of fructose consumption on the kidneys resembles its liver effect. Rats fed with a high-fructose diet exhibit bigger kidneys with higher triglycerides content, compared to control rats. The expression of ChREBPβ and its downstream genes was upregulated as well. Treating kidney-origin cells with fructose increased the expression of this factor as well, showing the direct effect of fructose on this factor. Thus, the appearance of fatty kidney in response to high-fructose consumption revealed a new mechanism linking metabolic syndrome to chronic kidney disease. Abstract Fructose consumption is associated with metabolic syndrome (MeS). Dysregulated lipid metabolism and ectopic lipid accumulation, such as in “fatty liver’’, are pivotal components of the syndrome. MeS is also associated with chronic kidney disease (CKD). The aim of this study was to evaluate kidney fructose metabolism and whether the addition of fructose leads to intrarenal fat accumulation. Sprague Dawley rats were fed either normal chow (Ctrl) or a high-fructose diet (HFrD). MeS features such as blood pressure and metabolic parameters in blood were measured. The kidneys were harvested for ChREBPβ and de novo lipogenesis (DNL) gene expression, triglyceride content and histopathology staining. HK2 (human kidney) cells were treated with fructose for 48 h and gene expression for ChREBPβ and DNL were determined. The HFrD rats exhibited higher blood pressure, glucose and triglyceride levels. The kidney weight of the HFrD rats was significantly higher than Ctrl rats. The difference can be explained by the higher triglyceride content in the HFrD kidneys. Oil red staining revealed lipid droplet formation in the HFrD kidneys, which was also supported by increased adipophilin mRNA expression. For ChREBPβ and its downstream genes, scd and fasn, mRNA expression was elevated in the HFrD kidneys. Treating HK2 cells with 40 mM fructose increased the expression of ChREBPβ. This study demonstrates that fructose consumption leads to intrarenal lipid accumulation and to the formation of a “fatty kidney”. This suggests a potential mechanism that can at least partially explain CKD development in fructose-induced MeS.
Collapse
Affiliation(s)
- Ariel Bier
- Medicine D, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel; (A.B.); (E.S.); (Y.S.); (E.G.)
| | - Eliyahu Shapira
- Medicine D, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel; (A.B.); (E.S.); (Y.S.); (E.G.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
| | - Rawan Khasbab
- Hypertension Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel;
| | - Yehonatan Sharabi
- Medicine D, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel; (A.B.); (E.S.); (Y.S.); (E.G.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
- Hypertension Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel;
| | - Ehud Grossman
- Medicine D, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel; (A.B.); (E.S.); (Y.S.); (E.G.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
- Hypertension Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel;
| | - Avshalom Leibowitz
- Medicine D, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel; (A.B.); (E.S.); (Y.S.); (E.G.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997801, Israel
- Hypertension Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan 5262000, Israel;
- Correspondence: ; Tel.: +972-35302834; Fax: +972-35302835
| |
Collapse
|
35
|
Na Z, Song J, Meng Y, Feng D, Wei J, Jiang H, Yang H, Peng Y, Cheng D, Fang Y, Li D. Serum fructose levels are positively correlated with dyslipidemia in women with polycystic ovary syndrome. Reprod Biomed Online 2022; 45:608-614. [DOI: 10.1016/j.rbmo.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/07/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022]
|
36
|
Therapeutic RNA-silencing oligonucleotides in metabolic diseases. Nat Rev Drug Discov 2022; 21:417-439. [PMID: 35210608 DOI: 10.1038/s41573-022-00407-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Recent years have seen unprecedented activity in the development of RNA-silencing oligonucleotide therapeutics for metabolic diseases. Improved oligonucleotide design and optimization of synthetic nucleic acid chemistry, in combination with the development of highly selective and efficient conjugate delivery technology platforms, have established and validated oligonucleotides as a new class of drugs. To date, there are five marketed oligonucleotide therapies, with many more in clinical studies, for both rare and common liver-driven metabolic diseases. Here, we provide an overview of recent developments in the field of oligonucleotide therapeutics in metabolism, review past and current clinical trials, and discuss ongoing challenges and possible future developments.
Collapse
|
37
|
Nishizawa H, Maeda N, Shimomura I. Impact of hyperuricemia on chronic kidney disease and atherosclerotic cardiovascular disease. Hypertens Res 2022; 45:635-640. [PMID: 35046512 DOI: 10.1038/s41440-021-00840-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
Hyperuricemia is caused by reduced renal/extrarenal excretion and overproduction of uric acid. It is affected by genetic predisposition related to uric acid transporters and by visceral fat accumulation due to overnutrition. The typical symptomatic complication of hyperuricemia is gout caused by monosodium urate crystals. Accumulated evidence from epidemiological studies suggests that hyperuricemia is also a risk factor for hypertension, chronic kidney disease (CKD) and atherosclerotic cardiovascular disease (CVD). However, it remains to be determined whether urate-lowering therapy for asymptomatic patients with hyperuricemia is effective in preventing CKD or CVD progression. This mini review focuses mainly on recent papers investigating the relationship between hyperuricemia and CKD or CVD and studies of urate-lowering therapy. Accumulated studies have proposed mechanisms of renal damage and atherosclerosis in hyperuricemia, including inflammasome activation, decreased nitric oxide bioavailability and oxidative stress induced by uric acid, urate crystals and xanthine oxidoreductase (XOR)-mediated reactive oxygen species. Since patients with hyperuricemia are a heterogeneous population with complex pathologies, it may be important to assess whether an outcome is the result of decreasing serum uric acid levels or an inhibitory effect on XOR. To clarify the impact of hyperuricemia on CKD and CVD progression, high-quality and detailed clinical and basic science studies of hyperuricemia and purine metabolism are needed.
Collapse
Affiliation(s)
- Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Norikazu Maeda
- Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
38
|
Mao D, Cheng J. Two dietary patterns from China might benefit kidney function, as indicated by latent profile analysis. J Ren Nutr 2022; 32:702-709. [DOI: 10.1053/j.jrn.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 11/29/2021] [Accepted: 01/10/2022] [Indexed: 11/11/2022] Open
|
39
|
Vallon V, Nakagawa T. Renal Tubular Handling of Glucose and Fructose in Health and Disease. Compr Physiol 2021; 12:2995-3044. [PMID: 34964123 PMCID: PMC9832976 DOI: 10.1002/cphy.c210030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The proximal tubule of the kidney is programmed to reabsorb all filtered glucose and fructose. Glucose is taken up by apical sodium-glucose cotransporters SGLT2 and SGLT1 whereas SGLT5 and potentially SGLT4 and GLUT5 have been implicated in apical fructose uptake. The glucose taken up by the proximal tubule is typically not metabolized but leaves via the basolateral facilitative glucose transporter GLUT2 and is returned to the systemic circulation or used as an energy source by distal tubular segments after basolateral uptake via GLUT1. The proximal tubule generates new glucose in metabolic acidosis and the postabsorptive phase, and fructose serves as an important substrate. In fact, under physiological conditions and intake, fructose taken up by proximal tubules is primarily utilized for gluconeogenesis. In the diabetic kidney, glucose is retained and gluconeogenesis enhanced, the latter in part driven by fructose. This is maladaptive as it sustains hyperglycemia. Moreover, renal glucose retention is coupled to sodium retention through SGLT2 and SGLT1, which induces secondary deleterious effects. SGLT2 inhibitors are new anti-hyperglycemic drugs that can protect the kidneys and heart from failing independent of kidney function and diabetes. Dietary excess of fructose also induces tubular injury. This can be magnified by kidney formation of fructose under pathological conditions. Fructose metabolism is linked to urate formation, which partially accounts for fructose-induced tubular injury, inflammation, and hemodynamic alterations. Fructose metabolism favors glycolysis over mitochondrial respiration as urate suppresses aconitase in the tricarboxylic acid cycle, and has been linked to potentially detrimental aerobic glycolysis (Warburg effect). © 2022 American Physiological Society. Compr Physiol 12:2995-3044, 2022.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, California, USA,Department of Pharmacology, University of California San Diego, La Jolla, California, USA,VA San Diego Healthcare System, San Diego, California, USA,Correspondence to and
| | - Takahiko Nakagawa
- Division of Nephrology, Rakuwakai-Otowa Hospital, Kyoto, Japan,Correspondence to and
| |
Collapse
|
40
|
Nakagawa T, Kang DH. Fructose in the kidney: from physiology to pathology. Kidney Res Clin Pract 2021; 40:527-541. [PMID: 34781638 PMCID: PMC8685370 DOI: 10.23876/j.krcp.21.138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022] Open
Abstract
The Warburg effect is a unique property of cancer cells, in which glycolysis is activated instead of mitochondrial respiration despite oxygen availability. However, recent studies found that the Warburg effect also mediates non-cancer disorders, including kidney disease. Currently, diabetes or glucose has been postulated to mediate the Warburg effect in the kidney, but it is of importance that the Warburg effect can be induced under nondiabetic conditions. Fructose is endogenously produced in several organs, including the kidney, under both physiological and pathological conditions. In the kidney, fructose is predominantly metabolized in the proximal tubules; under normal physiologic conditions, fructose is utilized as a substrate for gluconeogenesis and contributes to maintain systemic glucose concentration under starvation conditions. However, when present in excess, fructose likely becomes deleterious, possibly due in part to excessive uric acid, which is a by-product of fructose metabolism. A potential mechanism is that uric acid suppresses aconitase in the Krebs cycle and therefore reduces mitochondrial oxidation. Consequently, fructose favors glycolysis over mitochondrial respiration, a process that is similar to the Warburg effect in cancer cells. Activation of glycolysis also links to several side pathways, including the pentose phosphate pathway, hexosamine pathway, and lipid synthesis, to provide biosynthetic precursors as fuel for renal inflammation and fibrosis. We now hypothesize that fructose could be the mediator for the Warburg effect in the kidney and a potential mechanism for chronic kidney disease.
Collapse
Affiliation(s)
| | - Duk-Hee Kang
- Division of Nephrology, Department of Internal Medicine, Ewha Medical Research Institute, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
41
|
Pini N, Huo Z, Holland-Cunz S, Gros SJ. Increased Proliferation of Neuroblastoma Cells under Fructose Metabolism Can Be Measured by Isothermal Microcalorimetry. CHILDREN-BASEL 2021; 8:children8090784. [PMID: 34572216 PMCID: PMC8467942 DOI: 10.3390/children8090784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/28/2022]
Abstract
Neuroblastoma, like other cancer types, has an increased need for energy. This results in an increased thermogenic profile of the cells. How tumor cells optimize their energy efficiency has been discussed since Warburg described the fact that tumor cells prefer an anaerobic to an aerobic metabolism in the 1920s. An important question is how far the energy efficiency is influenced by the substrate. The aim of this study was to investigate how the metabolic activity of neuroblastoma cells is stimulated by addition of glucose or fructose to the medium and if this can be measured accurately by using isothermal microcalorimetry. Proliferation of Kelly and SH-EP Tet-21/N cells was determined in normal medium, in fructose-enriched, in glucose-enriched and in a fructose/glucose-enriched environment. Heat development of cells was measured by isothermal microcalorimetry. The addition of fructose, glucose or both to the medium led to increases in the metabolic activity of the cells, resulting in increased proliferation under the influence of fructose. These changes were reflected in an enhanced thermogenic profile, mirroring the results of the proliferation assay. The tested neuroblastoma cells prefer fructose metabolism over glucose metabolism, a quality that provides them with a survival benefit under unfavorable low oxygen and low nutrient supply when fructose is available. This can be quantified by measuring thermogenesis.
Collapse
Affiliation(s)
- Nicola Pini
- Department of Pediatric Surgery, University Children’s Hospital Basel, Spitalstr. 33, 4031 Basel, Switzerland; (N.P.); (Z.H.); (S.H.-C.)
- Department of Clinical Research, University of Basel, Schanzenstrasse 55, 4021 Basel, Switzerland
| | - Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, Spitalstr. 33, 4031 Basel, Switzerland; (N.P.); (Z.H.); (S.H.-C.)
- Department of Clinical Research, University of Basel, Schanzenstrasse 55, 4021 Basel, Switzerland
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, Spitalstr. 33, 4031 Basel, Switzerland; (N.P.); (Z.H.); (S.H.-C.)
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, Spitalstr. 33, 4031 Basel, Switzerland; (N.P.); (Z.H.); (S.H.-C.)
- Department of Clinical Research, University of Basel, Schanzenstrasse 55, 4021 Basel, Switzerland
- Correspondence:
| |
Collapse
|
42
|
Bian C, Wang Y, Li J, Gao J, Luan Z, Cui X, Ren H. Endogenous fructose is correlated with urinary albumin creatinine ratios and uric acid in type 2 diabetes mellitus. Diabetes Res Clin Pract 2021; 179:109034. [PMID: 34487756 DOI: 10.1016/j.diabres.2021.109034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/16/2021] [Accepted: 08/31/2021] [Indexed: 11/29/2022]
Abstract
AIM To detect the expression levels of fasting serum fructose and ketohexokinase (KHK) in patients with type 2 diabetes mellitus (T2DM) at different stages of urinary albumin creatinine ratios (UACR) and serum uric acid (sUA). METHODS 339 T2DM patients and 107 normal volunteers were divided into the normal uric acid (275 cases) and high uric acid group (171 cases) according to uric acid levels. T2DM patients were divided into the normal albuminuria group (118 cases, UACR < 30 mg/g), microalbuminuria group (112 cases, UACR 30-300 mg/g) and large amount of albuminuria group (109 cases, UACR > 300 mg/g). Levels of fasting serum fructose and KHK were detected and statistical analysis was carried out. RESULTS Fasting serum fructose and KHK levels increased with the increase of UACR and sUA (P < 0.05). Correlation analysis showed that fasting serum fructose and KHK levels were positively correlated with UACR and sUA (P < 0.05). Ridge regression analysis showed that fasting serum fructose and KHK were also correlated with urinary albumin and uric acid (P < 0.05). CONCLUSION Fasting serum fructose and KHK in endogenous fructose are associated with serum uric acid and urinary albumin levels in patients with T2DM. Trial number: ChiCTR2000039870.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology and Metabolism, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuxia Wang
- Department of Endocrinology and Metabolism, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jia Li
- Department of Endocrinology and Metabolism, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Gao
- Department of Gerontology, Xin Hua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaohui Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
43
|
Nakagawa T, Sanchez-Lozada LG, Andres-Hernando A, Kojima H, Kasahara M, Rodriguez-Iturbe B, Bjornstad P, Lanaspa MA, Johnson RJ. Endogenous Fructose Metabolism Could Explain the Warburg Effect and the Protection of SGLT2 Inhibitors in Chronic Kidney Disease. Front Immunol 2021; 12:694457. [PMID: 34220855 PMCID: PMC8243983 DOI: 10.3389/fimmu.2021.694457] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
Chronic low-grade inflammation underlies the pathogenesis of non-communicable diseases, including chronic kidney diseases (CKD). Inflammation is a biologically active process accompanied with biochemical changes involving energy, amino acid, lipid and nucleotides. Recently, glycolysis has been observed to be increased in several inflammatory disorders, including several types of kidney disease. However, the factors initiating glycolysis remains unclear. Added sugars containing fructose are present in nearly 70 percent of processed foods and have been implicated in the etiology of many non-communicable diseases. In the kidney, fructose is transported into the proximal tubules via several transporters to mediate pathophysiological processes. Fructose can be generated in the kidney during glucose reabsorption (such as in diabetes) as well as from intra-renal hypoxia that occurs in CKD. Fructose metabolism also provides biosynthetic precursors for inflammation by switching the intracellular metabolic profile from mitochondrial oxidative phosphorylation to glycolysis despite the availability of oxygen, which is similar to the Warburg effect in cancer. Importantly, uric acid, a byproduct of fructose metabolism, likely plays a key role in favoring glycolysis by stimulating inflammation and suppressing aconitase in the tricarboxylic acid cycle. A consequent accumulation of glycolytic intermediates connects to the production of biosynthetic precursors, proteins, lipids, and nucleic acids, to meet the increased energy demand for the local inflammation. Here, we discuss the possibility of fructose and uric acid may mediate a metabolic switch toward glycolysis in CKD. We also suggest that sodium-glucose cotransporter 2 (SGLT2) inhibitors may slow the progression of CKD by reducing intrarenal glucose, and subsequently fructose levels.
Collapse
Affiliation(s)
- Takahiko Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, Kyoto, Japan.,Department of Biochemistry, Shiga University of Medical Science, Otsu, Japan
| | - Laura G Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, United States
| | - Hideto Kojima
- Department of Biochemistry, Shiga University of Medical Science, Otsu, Japan
| | - Masato Kasahara
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Bernardo Rodriguez-Iturbe
- Department of Nephrology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran and Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
| | - Petter Bjornstad
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, United States.,Department of Pediatrics-Endocrinology, University of Colorado Denver, Aurora, CO, United States
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, United States
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
44
|
Shi YN, Liu YJ, Xie Z, Zhang WJ. Fructose and metabolic diseases: too much to be good. Chin Med J (Engl) 2021; 134:1276-1285. [PMID: 34010200 PMCID: PMC8183764 DOI: 10.1097/cm9.0000000000001545] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Excessive consumption of fructose, the sweetest of all naturally occurring carbohydrates, has been linked to worldwide epidemics of metabolic diseases in humans, and it is considered an independent risk factor for cardiovascular diseases. We provide an overview about the features of fructose metabolism, as well as potential mechanisms by which excessive fructose intake is associated with the pathogenesis of metabolic diseases both in humans and rodents. To accomplish this aim, we focus on illuminating the cellular and molecular mechanisms of fructose metabolism as well as its signaling effects on metabolic and cardiovascular homeostasis in health and disease, highlighting the role of carbohydrate-responsive element-binding protein in regulating fructose metabolism.
Collapse
Affiliation(s)
- Ya-Nan Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Ya-Jin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
| | - Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Weiping J. Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin 300134, China
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
45
|
Woodhams L, Sim TF, Chalmers L, Yeap B, Green D, Schlaich M, Schultz C, Hillis G. Diabetic kidney disease in type 2 diabetes: a review of pathogenic mechanisms, patient-related factors and therapeutic options. PeerJ 2021; 9:e11070. [PMID: 33976959 PMCID: PMC8061574 DOI: 10.7717/peerj.11070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/16/2021] [Indexed: 12/23/2022] Open
Abstract
The global prevalence of diabetic kidney disease is rapidly accelerating due to an increasing number of people living with type 2 diabetes. It has become a significant global problem, increasing human and financial pressures on already overburdened healthcare systems. Interest in diabetic kidney disease has increased over the last decade and progress has been made in determining the pathogenic mechanisms and patient-related factors involved in the development and pathogenesis of this disease. A greater understanding of these factors will catalyse the development of novel treatments and influence current practice. This review summarises the latest evidence for the factors involved in the development and progression of diabetic kidney disease, which will inform better management strategies targeting such factors to improve therapeutic outcomes in patients living with diabetes.
Collapse
Affiliation(s)
- Louise Woodhams
- Curtin Medical School, Curtin University of Technology, Perth, Western Australia, Australia
| | - Tin Fei Sim
- Curtin Medical School, Curtin University of Technology, Perth, Western Australia, Australia
| | - Leanne Chalmers
- Curtin Medical School, Curtin University of Technology, Perth, Western Australia, Australia
| | - Bu Yeap
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Western Australia, Australia.,Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Daniel Green
- School of Human Sciences (Exercise and Sport Sciences), The University of Western Australia, Perth, Western Australia, Australia
| | - Markus Schlaich
- Medical School, The University of Western Australia, Perth, Western Australia, Australia.,Department of Cardiology and Nephrology, Royal Perth Hospital, Perth, Western Australia, Australia.,Neurovascular Hypertension and Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, Western Australia, Australia
| | - Carl Schultz
- Medical School, The University of Western Australia, Perth, Western Australia, Australia.,Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Graham Hillis
- Medical School, The University of Western Australia, Perth, Western Australia, Australia.,Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
46
|
Carminic acid supplementation protects against fructose-induced kidney injury mainly through suppressing inflammation and oxidative stress via improving Nrf-2 signaling. Aging (Albany NY) 2021; 13:10326-10353. [PMID: 33819919 PMCID: PMC8064181 DOI: 10.18632/aging.202794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
Excessive fructose (Fru) intake has become an increased risk for chronic kidney disease progression. Despite extensive researches that have been performed to develop effective treatments against Fru-induced renal injury, the outcome has achieved limited success. In this study, we attempted to explore whether carminic acid (CA) could influence the progression of Fru-induced kidney injury, and the underlying molecular mechanism. At first, our in vitro results showed that CA significantly reduced inflammation in mouse tubular epithelial cells and human tubule epithelial cells stimulated by Fru. The anti-inflammatory effects of CA were associated with the blockage of nuclear factor-κB (NF-κB) signaling. In addition, Fru-exposed cells showed higher oxidative stress, which was effectively restrained by CA treatment through improving nuclear factor (erythroid-derived 2)-like 2 (Nrf-2) nuclear translocation. Importantly, we found that Fru-induced inflammation and oxidative stress were accelerated in cells with Nrf-2 knockdown. What's more, in Fru-stimulated cells, CA-alleviated inflammatory response and reactive oxygen species (ROS) production were evidently abolished by Nrf-2 knockdown. The in vivo analysis demonstrated that Fru led to metabolic disorder, excessive albuminuria and histologic changes in renal tissues, which were effectively reversed by CA supplementation. We confirmed that CA significantly reduced inflammation and oxidative stress in the kidneys of mice through regulating NF-κB and Nrf-2 signaling pathways, eventually alleviating the progression of chronic kidney injury. Taken together, these results identified CA as a potential therapeutic strategy for metabolic stress-induced renal injury through restraining inflammation and oxidative stress via the improvement of Nrf-2 signaling.
Collapse
|
47
|
Watanabe K, Nakayama M, Yamamoto T, Yamada G, Sato H, Miyazaki M, Ito S. Different clinical impact of hyperuricemia according to etiologies of chronic kidney disease: Gonryo Study. PLoS One 2021; 16:e0249240. [PMID: 33765101 PMCID: PMC7993817 DOI: 10.1371/journal.pone.0249240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/15/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Hyperuricemia is highly prevalent in chronic kidney disease (CKD) patients, but the evidence for a relationship between uric acid (UA) and clinical outcomes in CKD patients is limited and inconsistent. We hypothesized that UA has a different impact on clinical outcomes according to the underlying disease causing CKD. METHODS This study prospectively investigated the associations between UA and renal and non-renal outcomes according to the underlying disease causing CKD in 2,797 Japanese patients under the care of nephrologists. The patients were categorized into four groups: primary renal disease (n = 1306), hypertensive nephropathy (n = 467), diabetic nephropathy (n = 275), and other nephropathy (n = 749). The renal outcome was defined as end-stage renal disease (ESRD), and the non-renal outcome was defined as a composite endpoint of cardiovascular events (CVEs) and all-cause mortality. RESULTS During a median 4.8-year follow-up, 359 (12.8%) patients reached the renal outcome, and 260 (9.3%) reached the non-renal outcome. In the all-patient analysis, hyperuricemia was not associated with the risks for renal and non-renal outcomes, but in primary renal disease (PRD) and hypertensive renal disease (HTN) patients, hyperuricemia was significantly associated with non-renal outcomes. Per 1 mg/dl higher UA level, multivariable adjusted hazard ratio was 1.248 (95% CI: 1.003 to 1.553) for PRD, and 1.250 (1.035 to 1.510) for HTN. Allopurinol did not reduce the risks for renal and non-renal outcomes, both in all patients and in the subgroup analysis. CONCLUSIONS The effect of hyperuricemia on clinical outcomes in CKD patients varies according to the underlying disease causing CKD. Hyperuricemia is an independent risk factor for non-renal outcomes in primary renal disease and hypertensive renal disease patients. Allopurinol did not decrease the risks for renal and non-renal outcomes.
Collapse
Affiliation(s)
- Kimio Watanabe
- Division of Kidney Center, St Luke’s International Hospital, Tokyo, Japan
| | - Masaaki Nakayama
- Division of Kidney Center, St Luke’s International Hospital, Tokyo, Japan
- Research Division of Chronic Kidney Disease and Dialysis Treatment, Tohoku University Hospital, Sendai, Japan
| | - Tae Yamamoto
- Division of Kidney Center, Sendai City Hospital, Sendai, Japan
| | - Gen Yamada
- Division of Nephrology and Endocrinology, Osaki Citizen Hospital, Osaki, Japan
| | - Hiroshi Sato
- Division of Internal Medicine, JR Sendai Hospital, Sendai, Japan
| | - Mariko Miyazaki
- Division of Blood Purification, Tohoku University Hospital, Sendai, Japan
- Division of Nephrology, Hypertension and Endocrinology, Tohoku University, Sendai, Japan
| | - Sadayoshi Ito
- Division of Nephrology, Hypertension and Endocrinology, Tohoku University, Sendai, Japan
- Katta General Public Hospital, Shiroishi, Japan
| |
Collapse
|
48
|
Zhang C, Li L, Zhang Y, Zeng C. Recent advances in fructose intake and risk of hyperuricemia. Biomed Pharmacother 2020; 131:110795. [PMID: 33152951 DOI: 10.1016/j.biopha.2020.110795] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/17/2022] Open
Abstract
With the widespread popularity of hyperuricemia, it has become a severe threat to human public health. Accumulating evidence suggests that dietary fructose has a close relationship with hyperuricemia, but the role of fructose intake in hyperuricemia remains unclear. Hyperuricemia is characterized by excessive production and deposition of urate crystals. Metabolism of fructose leads to the increased serum concentration of urate. In this review, we depict an update of fructose consumption worldwide and the epidemiology of hyperuricemia and summarize the progress in studying the relationship between fructose intake and the risk of hyperuricemia. This review highlights the metabolic process of fructose in the liver, small intestine, and kidney. Furthermore, we discuss molecular insights on fructose metabolism to reveal the underlying mechanism of fructose metabolism. Additionally, we elaborate on the effect of fructose metabolism on hyperuricemia to deeply understand the pathogenesis of hyperuricemia caused by fructose intake. Fructose consumption has a close correlation with an enhanced risk of developing hyperuricemia. More prospective studies are inevitable to understand the role of fructose intake in the development of hyperuricemia.
Collapse
Affiliation(s)
- Congwang Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China
| | - Lijun Li
- Department of Quality Control, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China
| | - Yipeng Zhang
- Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, 518110, PR China.
| |
Collapse
|
49
|
Nakagawa T, Lanaspa MA, Millan IS, Fini M, Rivard CJ, Sanchez-Lozada LG, Andres-Hernando A, Tolan DR, Johnson RJ. Fructose contributes to the Warburg effect for cancer growth. Cancer Metab 2020; 8:16. [PMID: 32670573 PMCID: PMC7350662 DOI: 10.1186/s40170-020-00222-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity and metabolic syndrome are strongly associated with cancer, and these disorders may share a common mechanism. Recently, fructose has emerged as a driving force to develop obesity and metabolic syndrome. Thus, we assume that fructose may be the mechanism to explain why obesity and metabolic syndrome are linked with cancer. Clinical and experimental evidence showed that fructose intake was associated with cancer growth and that fructose transporters are upregulated in various malignant tumors. Interestingly, fructose metabolism can be driven under low oxygen conditions, accelerates glucose utilization, and exhibits distinct effects as compared to glucose, including production of uric acid and lactate as major byproducts. Fructose promotes the Warburg effect to preferentially downregulate mitochondrial respiration and increases aerobic glycolysis that may aid metastases that initially have low oxygen supply. In the process, uric acid may facilitate carcinogenesis by inhibiting the TCA cycle, stimulating cell proliferation by mitochondrial ROS, and blocking fatty acid oxidation. Lactate may also contribute to cancer growth by suppressing fat oxidation and inducing oncogene expression. The ability of fructose metabolism to directly stimulate the glycolytic pathway may have been protective for animals living with limited access to oxygen, but may be deleterious toward stimulating cancer growth and metastasis for humans in modern society. Blocking fructose metabolism may be a novel approach for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Takahiko Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, 2 Otowa-Chinji-cho, Yamashina-ku, Kyoto, Japan
- Department of Stem Cell Biology & Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Inigo San Millan
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, USA
| | - Mehdi Fini
- University of Colorado Cancer Center, Aurora, CO USA
| | | | - Laura G. Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, 14080 Mexico City, CP Mexico
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Dean R. Tolan
- Department of Biology, Boston University, Boston, MA USA
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| |
Collapse
|
50
|
Affiliation(s)
- Daniel I Feig
- From the Department of Pediatrics, Division of Pediatric Nephrology, University of Alabama School of Medicine, Birmingham
| |
Collapse
|