1
|
Chen Z, Malek V, Natarajan R. Update: the role of epigenetics in the metabolic memory of diabetic complications. Am J Physiol Renal Physiol 2024; 327:F327-F339. [PMID: 38961840 PMCID: PMC11460341 DOI: 10.1152/ajprenal.00115.2024] [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: 04/12/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
Diabetes, a chronic disease characterized by hyperglycemia, is associated with significantly accelerated complications, including diabetic kidney disease (DKD), which increases morbidity and mortality. Hyperglycemia and other diabetes-related environmental factors such as overnutrition, sedentary lifestyles, and hyperlipidemia can induce epigenetic changes. Working alone or with genetic factors, these epigenetic changes that occur without alterations in the underlying DNA sequence, can alter the expression of pathophysiological genes and impair functions of associated target cells/organs, leading to diabetic complications like DKD. Notably, some hyperglycemia-induced epigenetic changes persist in target cells/tissues even after glucose normalization, leading to sustained complications despite glycemic control, so-called metabolic memory. Emerging evidence from in vitro and in vivo animal models and clinical trials with subjects with diabetes identified clear associations between metabolic memory and epigenetic changes including DNA methylation, histone modifications, chromatin structure, and noncoding RNAs at key loci. Targeting such persistent epigenetic changes and/or molecules regulated by them can serve as valuable opportunities to attenuate, or erase metabolic memory, which is crucial to prevent complication progression. Here, we review these cell/tissue-specific epigenetic changes identified to-date as related to diabetic complications, especially DKD, and the current status on targeting epigenetics to tackle metabolic memory. We also discuss limitations in current studies, including the need for more (epi)genome-wide studies, integrative analysis using multiple epigenetic marks and Omics datasets, and mechanistic evaluation of metabolic memory. Considering the tremendous technological advances in epigenomics, genetics, sequencing, and availability of genomic datasets from clinical cohorts, this field is likely to see considerable progress in the upcoming years.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, California
| | - Vajir Malek
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, California
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, California
| |
Collapse
|
2
|
P. Simões D, Moreira Perez M, Aguiar Alves BDC, Araújo Encinas JF, Santos Raimundo JR, Costas Arcia CG, Lopes Mathia V, Sacchi Mendonça MI, Mesiano Maifrino LB, Murad N, Affonso Fonseca FL, Luciano da Veiga G. A Cross-Sectional Study of p66Shc Gene Expression in Liquid Biopsy of Diabetic Patients. Is it Possible to Predict the Onset of Renal Disease? INTERNATIONAL JOURNAL OF MEDICAL STUDENTS 2023. [DOI: 10.5195/ijms.2022.1306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Background: Diabetic nephropathy (DN) is a disorder affecting glomerular function that, histologically, is due to the presence of glomerulosclerosis accompanied with endothelial dysfunction of the afferent and efferent renal arterioles. Insulin resistance in diabetic patients is known to be one of the causes of endothelial dysfunction because it increases oxidative stress, and one of the main genes regulating the production pathways of reactive oxygen species is p66Shc. The aim of this study was to evaluate the p66Shc gene expression as a precocious biomarker of renal dysfunction in diabetic patients, using liquids samples of urine sediment and peripheral blood.
Methods: 29 diabetic patients and 37 healthy donors were recruited from the Centro Universitário FMABC outpatient clinic. The RT-gPCR technique was applied to evaluate p66Shc gene expression in urine and peripheral blood samples from diabetic patients, which were compared with healthy donors.
Results: There was no significant expression of p66Shc gene in samples from diabetic patients compared with healthy donors. However, p66Shc expression in the blood samples of diabetics (0.02417±0.078652-ΔCT, n=29) was 3.6 times higher than in healthy participants (0.00689±0.01758, n=37) while in the urine samples, it was 1.48 times higher in diabetics group (0.02761±0.05412-ΔCT) than in CTL group (0.0186±0.02199).
Conclusion: There was no significant p66Shc gene expression in peripheral blood and urine samples of diabetic patients without kidney injury compared with healthy donors, although there is a tendency for this gene to participate in the oxidative imbalance present in diabetes.
Collapse
|
3
|
Song Y, Yu H, Sun Q, Pei F, Xia Q, Gao Z, Li X. Grape seed proanthocyanidin extract targets p66Shc to regulate mitochondrial biogenesis and dynamics in diabetic kidney disease. Front Pharmacol 2023; 13:1035755. [PMID: 36686673 PMCID: PMC9853208 DOI: 10.3389/fphar.2022.1035755] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
Mitochondrial biogenesis and dynamics are associated with renal mitochondrial dysfunction and the pathophysiological development of diabetic kidney disease (DKD). Decreased p66Shc expression prevents DKD progression by significantly regulating mitochondrial function. Grape seed proanthocyanidin extract (GSPE) is a potential therapeutic medicine for multiple kinds of diseases. The effect of GSPE on the mitochondrial function and p66Shc in DKD has not been elucidated. Hence, we decided to identify p66Shc as a therapeutic target candidate to probe whether GSPE has a renal protective effect in DKD and explored the underlying mechanisms. METHODS In vivo, rats were intraperitoneally injected with streptozotocin (STZ) and treated with GSPE. Biochemical changes, mitochondrial morphology, the ultrastructure of nephrons, and protein expression of mitochondrial biogenesis (SIRT1, PGC-1α, NRF1, TFAM) and dynamics (DRP1, MFN1) were determined. In vitro, HK-2 cells were transfected with p66Shc and treated with GSPE to evaluate changes in cell apoptosis, reactive oxygen species (ROS), mitochondrial quality, the protein expression. RESULTS In vivo, GSPE significantly improved the renal function of rats, with less proteinuria and a lower apoptosis rate in the injured renal tissue. Besides, GSPE treatment increased SIRT1, PGC-1α, NRF1, TFAM, and MFN1 expression, decreased p66Shc and DRP1 expression. In vitro, overexpression of p66Shc decreased the resistance of HK-2 cells to high glucose toxicity, as shown by increased apoptosis and ROS production, decreased mitochondrial quality and mitochondrial biogenesis, and disturbed mitochondrial dynamic homeostasis, ultimately leading to mitochondrial dysfunction. While GSPE treatment reduced p66Shc expression and reversed these changes. CONCLUSION GSPE can maintain the balance between mitochondrial biogenesis and dynamics by negatively regulating p66Shc expression.
Collapse
Affiliation(s)
- Yiyun Song
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Yu
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qiaoling Sun
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fei Pei
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qing Xia
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhaoli Gao
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China,Department of Nephrology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, China,*Correspondence: Zhaoli Gao, ; Xianhua Li,
| | - Xianhua Li
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China,Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China,*Correspondence: Zhaoli Gao, ; Xianhua Li,
| |
Collapse
|
4
|
Mousavi S, Khazeei Tabari MA, Bagheri A, Samieefar N, Shaterian N, Kelishadi R. The Role of p66Shc in Diabetes: A Comprehensive Review from Bench to Bedside. J Diabetes Res 2022; 2022:7703520. [PMID: 36465704 PMCID: PMC9715346 DOI: 10.1155/2022/7703520] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
It is well-documented that diabetes is an inflammatory and oxidative disease, with an escalating global burden. Still, there is no definite treatment for diabetes or even prevention of its harmful complications. Therefore, understanding the molecular pathways associated with diabetes might help in finding a solution. p66Shc is a member of Shc family proteins, and it is considered as an oxidative stress sensor and regulator in cells. There are inconsistent data about the role of p66Shc in inducing diabetes, but accumulating evidence supports its role in the pathogenesis of diabetes-related complications, including macro and microangiopathies. There is growing hope that by understanding and targeting molecular pathways involved in this network, prevention of diabetes or its complications would be achievable. This review provides an overview about the role of p66Shc in the development of diabetes and its complications.
Collapse
Affiliation(s)
- SeyedehFatemeh Mousavi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Mazandaran, Iran
- USERN Office, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Alireza Bagheri
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Noosha Samieefar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Shaterian
- Student Research Committee, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- USERN Office, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Roya Kelishadi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- USERN Office, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Lee DY, Kim JY, Ahn E, Hyeon JS, Kim GH, Park KJ, Jung Y, Lee YJ, Son MK, Kim SW, Han SY, Kim JH, Roh GS, Cha DR, Hwang GS, Kim WH. Associations between local acidosis induced by renal LDHA and renal fibrosis and mitochondrial abnormalities in patients with diabetic kidney disease. Transl Res 2022; 249:88-109. [PMID: 35788054 DOI: 10.1016/j.trsl.2022.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/23/2022] [Accepted: 06/22/2022] [Indexed: 10/31/2022]
Abstract
During the progression of diabetic kidney disease (DKD), renal lactate metabolism is rewired. The relationship between alterations in renal lactate metabolism and renal fibrosis in patients with diabetes has only been partially established due to a lack of biopsy tissues from patients with DKD and the intricate mechanism of lactate homeostasis. The role of lactate dehydrogenase A (LDHA)-mediated lactate generation in renal fibrosis and dysfunction in human and animal models of DKD was explored in this study. Measures of lactate metabolism (urinary lactate levels and LDHA expression) and measures of DKD progression (estimated glomerular filtration rate and Wilms' tumor-1 expression) were strongly negatively correlated in patients with DKD. Experiments with streptozotocin-induced DKD rat models and the rat renal mesangial cell model confirmed our findings. We found that the pathogenesis of DKD is linked to hypoxia-mediated lactic acidosis, which leads to fibrosis and mitochondrial abnormalities. The pathogenic characteristics of DKD were significantly reduced when aerobic glycolysis or LDHA expression was inhibited. Further studies will aim to investigate whether local acidosis caused by renal LDHA might be exploited as a therapeutic target in patients with DKD.
Collapse
Affiliation(s)
- Dae-Yeon Lee
- Division of Cardiovascular Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea; Department of Anatomy and Convergence Medical Science, Bio Anti-aging Medical Research Center, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Ji-Yeon Kim
- Division of Cardiovascular Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Eunyong Ahn
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Jin Seong Hyeon
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Gyu-Hee Kim
- Division of Metabolic Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Keon-Jae Park
- Division of Metabolic Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Youngae Jung
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Yoo-Jeong Lee
- Division of Metabolic Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Mi Kyoung Son
- Division of Cardiovascular Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Seung Woo Kim
- Division of Cardiovascular Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Sang Youb Han
- Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Republic of Korea
| | - Jae-Hong Kim
- Division of Life Sciences, College of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Bio Anti-aging Medical Research Center, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Dae Ryong Cha
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea.
| | - Geum-Sook Hwang
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea.
| | - Won-Ho Kim
- Division of Cardiovascular Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea.
| |
Collapse
|
6
|
Zhao C, Li L, Li C, Tang C, Cai J, Liu Y, Yang J, Xi Y, Yang M, Jiang N, Han Y, Liu Y, Luo S, Xiao L, Sun L. PACS-2 deficiency in tubular cells aggravates lipid-related kidney injury in diabetic kidney disease. Mol Med 2022; 28:117. [PMID: 36138342 PMCID: PMC9502582 DOI: 10.1186/s10020-022-00545-x] [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: 04/13/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background Lipid accumulation in tubular cells plays a key role in diabetic kidney disease (DKD). Targeting lipid metabolism disorders has clinical value in delaying the progression of DKD, but the precise mechanism by which molecules mediate lipid-related kidney injury remains unclear. Phosphofurin acidic cluster sorting protein 2 (PACS-2) is a multifunctional sorting protein that plays a role in lipid metabolism. This study determined the role of PACS-2 in lipid-related kidney injury in DKD. Methods Diabetes was induced by a high-fat diet combined with intraperitoneal injections of streptozotocin (HFD/STZ) in proximal tubule-specific knockout of Pacs-2 mice (PT-Pacs-2−/− mice) and the control mice (Pacs-2fl/fl mice). Transcriptomic analysis was performed between Pacs-2fl/fl mice and PT-Pacs-2−/− mice. Results Diabetic PT-Pacs-2−/− mice developed more severe tubule injury and proteinuria compared to diabetic Pacs-2fl/fl mice, which accompanied with increasing lipid synthesis, uptake and decreasing cholesterol efflux as well as lipid accumulation in tubules of the kidney. Furthermore, transcriptome analysis showed that the mRNA level of sterol O-acyltransferase 1 (Soat1) was up-regulated in the kidney of control PT-Pacs-2−/− mice. Transfection of HK2 cells with PACS-2 siRNA under high glucose plus palmitic acid (HGPA) condition aggravated lipid deposition and increased the expression of SOAT1 and sterol regulatory element-binding proteins (SREBPs), while the effect was blocked partially in that of co-transfection of SOAT1 siRNA. Conclusions PACS-2 has a protective role against lipid-related kidney injury in DKD through SOAT1/SREBPs signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00545-x.
Collapse
Affiliation(s)
- Chanyue Zhao
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Li Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Chenrui Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yu Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Jinfei Yang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yiyun Xi
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Ming Yang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Na Jiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yachun Han
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Yan Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Shilu Luo
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Li Xiao
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Lin Sun
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
7
|
Li Y, Ou S, Liu Q, Gan L, Zhang L, Wang Y, Qin J, Liu J, Wu W. Genistein improves mitochondrial function and inflammatory in rats with diabetic nephropathy via inhibiting MAPK/NF-κB pathway. Acta Cir Bras 2022; 37:e370601. [PMID: 35976278 PMCID: PMC9377651 DOI: 10.1590/acb370601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/20/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose: To investigate the effect of genistein on inflammation and mitochondrial function of diabetic nephropathy. Methods: Diabetic nephropathy model was established in Sprague-Dawley rats. Automatic biochemical analyzer was employed to detect the kidney function index, serum creatinine, serum urea nitrogen, and 24 h-urine protein and blood glucose. Hematoxylin and eosin staining and periodic acid Schiff staining were used to observe renal morphology. Mitochondrial changes and podocyte integrity were monitored by transmission electron microscope. The expression levels of mfn2, NOX4, P53, MAPK, and NF-κB were detected by Western blotting. The changes of mitochondrial membrane potential were measured by JC-1. The level of mfn2 was assessed by immunofluorescence assay. Results: Genistein ameliorated the kidney function with reduced Scr and blood glucose. The expressions of NOX4, MAPK, p65 and p53 were downregulated, while the expression of mnf2 was the opposite in genistein-treated kidneys. Further investigations revealed that genistein reduced expansion of mesangial matrix and oxidative stress, protected podocyte integrity and increased mitochondrial membrane potential. Conclusions: Genistein could alleviate diabetic nephropathy through inhibiting MAPK/NF-κB pathway, improving mitochondrial function and anti-inflammatory.
Collapse
Affiliation(s)
- Ying Li
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Santao Ou
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Qi Liu
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Linwang Gan
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Liling Zhang
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Yujie Wang
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Jianhua Qin
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Jin Liu
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| | - Weihua Wu
- MD. SiChuan Clinical Research Center for Nephropathy - Affiliated Hospital of Southwest Medical University - Department of Nephrology - Luzhou, China
| |
Collapse
|
8
|
Xu W, Zhang H, Zhang Q, Xu J. β-Amyrin ameliorates diabetic nephropathy in mice and regulates the miR-181b-5p/HMGB2 axis in high glucose-stimulated HK-2 cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:637-649. [PMID: 34894065 DOI: 10.1002/tox.23431] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/22/2021] [Accepted: 11/27/2021] [Indexed: 06/14/2023]
Abstract
Diabetic nephropathy (DN) is a diabetic complication that can cause renal failure. β-amyrin has been identified to possess anti-diabetic property. This study was designed to evaluate the potential role of β-amyrin in DN and its underlying mechanism. Streptozotocin-induced diabetic mice were used as the in vivo model, and high glucose (HG)-stimulated human proximal tubular HK-2 cells were utilized as the in vitro model. Renal histological changes in mice were assessed by hematoxylin-eosin and periodic acid-Schiff staining. HK-2 cell viability and apoptosis were detected by Cell Counting Kit-8 assay and flow cytometry analysis, respectively. β-amyrin was found to ameliorate kidney injury in DN mice and suppressed inflammatory response as well as apoptosis of HG-stimulated HK-2 cells. miR-181-5p expression in murine renal tissues and HK-2 cells was detected by in situ hybridization (ISH) and fluorescence in situ hybridization (FISH). MiR-181b-5p, a previously identified target for diabetic kidney disease, was downregulated in renal tissues and HG stimulated HK-2 cells, and β-amyrin induced the upregulation of miR-181b-5p. Binding relationship between miR-181b-5p and high mobility group box 2 (HMGB2) was confirmed by luciferase reporter assay. MiR-181b-5p bound to 3' untranslated region of HMGB2 to suppress its expression. As shown by immunohistochemical staining and immunofluorescence staining, HMGB2 was upregulated in the in vivo and in vitro models of DN, and β-amyrin induced the downregulation of HMGB2. Moreover, HMGB2 overexpression neutralized the suppressive effects of miR-181b-5p elevation on the inflammatory response and apoptosis of HG-treated HK-2 cells. Overall, β-amyrin ameliorates DN in mice and suppresses inflammatory response and apoptosis of HG-stimulated HK-2 cells via the miR-181b-5p/HMGB2 axis.
Collapse
Affiliation(s)
- Wenhua Xu
- Preventive Treatment Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Hongwu Zhang
- Department of Endocrinology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Qinfeng Zhang
- Department of Endocrinology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Jialan Xu
- Preventive Treatment Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Xing L, Wu S, Shi Y, Yue F, Wei L, Russell R, Zhang D. Chronic constant light exposure aggravates high fat diet-induced renal injury in rats. Front Endocrinol (Lausanne) 2022; 13:900392. [PMID: 35966094 PMCID: PMC9372432 DOI: 10.3389/fendo.2022.900392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity-related kidney disease is now recognized as a global health issue, with a substantial number of patients developing progressive renal failure and end-stage renal disease. Interestingly, recent studies indicate light pollution is a novel environmental risk factor for chronic kidney disease. However, the impact of light pollution on obesity-related kidney disease remains largely unknown, with its underlying mechanism insufficiently explained. Renal hypoxia induced factor 1α (HIF1α) is critical in the development of glomerulosclerosis and renal fibrosis. The present study explored effects of constant light exposure on high fat diet (HFD) -induced renal injury and its association with HIF1α signal pathway. Thirty-two male Sprague Dawley rats were divided into four groups according to diet (HFD or normal chow diet) and light cycles (light/dark or constant light). After 16 weeks treatment, rats were sacrificed and pathophysiological assessments were performed. In normal chow fed rats, constant light exposure led to glucose abnormalities and dyslipidemia. In HFD fed rats, constant light exposure exacerbated obesity, glucose abnormalities, insulin resistance, dyslipidemia, renal functional decline, proteinuria, glomerulomegaly, renal inflammation and fibrosis. And, constant light exposure caused an increase in HIF1α and a decrease in prolyl hydroxylase domain 1 (PHD1) and PHD2 expression in kidneys of HFD-fed rats. Then, we demonstrated that BMAL1 bound directly to the promoters of PHD1 in mouse podocyte clone 5 cell line (MPC5) by ChIP assays. In conclusion, chronic constant light exposure aggravates HFD-induced renal injuries in rats, and it is associated with activation of HIF1α signal pathway.
Collapse
Affiliation(s)
- Lin Xing
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanyu Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Fangzhi Yue
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Wei
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Ryan Russell
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Dongmei Zhang,
| |
Collapse
|
10
|
Salami M, Salami R, Mafi A, Aarabi MH, Vakili O, Asemi Z. Therapeutic potential of resveratrol in diabetic nephropathy according to molecular signaling. Curr Mol Pharmacol 2021; 15:716-735. [PMID: 34923951 DOI: 10.2174/1874467215666211217122523] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Diabetic nephropathy (DN) as a severe complication of diabetes mellitus (DM), is a crucial menace for human health and survival and remarkably elevates the healthcare systems' costs. Therefore, it is worth noting to identify novel preventive and therapeutic strategies to alleviate the disease conditions. Resveratrol, as a well-defined anti-diabetic/ antioxidant agent has capabilities to counteract diabetic complications. It has been predicted that resveratrol will be a fantastic natural polyphenol for diabetes therapy in the next few years. OBJECTIVE Accordingly, the current review aims to depict the role of resveratrol in the regulation of different signaling pathways that are involved in the reactive oxygen species (ROS) production, inflammatory processes, autophagy, and mitochondrial dysfunction, as critical contributors to DN pathophysiology. RESULTS The pathogenesis of DN can be multifactorial; hyperglycemia is one of the prominent risk factors of DN development that is closely related to oxidative stress. Resveratrol, as a well-defined polyphenol, has various biological and medicinal properties, including anti-diabetic, anti-inflammatory, and anti-oxidative effects. CONCLUSION Resveratrol prevents kidney damages that are caused by oxidative stress, enhances antioxidant capacity, and attenuates the inflammatory and fibrotic responses. For this reason, resveratrol is considered an interesting target in DN research due to its therapeutic possibilities during diabetic disorders and renal protection.
Collapse
Affiliation(s)
- Marziyeh Salami
- Department of biochemistry, Faculty of medicine, Semnan University of medical sciences, Semnan, Iran
| | - Raziyeh Salami
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad-Hossein Aarabi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
11
|
Jelinek HF, Helf C, Khalaf K. Human SHC-transforming protein 1 and its isoforms p66shc: A novel marker for prediabetes. J Diabetes Investig 2021; 12:1881-1889. [PMID: 33759377 PMCID: PMC8504898 DOI: 10.1111/jdi.13551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 03/02/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Prediabetes is a multifactorial condition. Current guidelines for diabetes screening recommend either the use of glycated hemoglobin (HbA1c), or blood glucose level (BGL). This research aimed to identify if p66shc a component of the Human SHC-Transforming Protein 1 (Shc1), a mitochondrial associated oxidative stress biomarker, is significantly altered in patients with elevated BGL. Furthermore, we evaluated if inflammatory and oxidative stress markers, such as p66shc, are a useful addition to the regularly used biomarkers to increase sensitivity for identification of prediabetes. METHODS All participants attended the Diabetic Health Screening at Charles Sturt University (CSU), Australia. The cross-sectional clinical study collected demographic and clinical variables from 346 participants and classified into control or prediabetes based on fasting BGL. Blood and urine samples were analyzed for oxidative stress and inflammation markers. Logistic regression was used to compare multidimensional diagnostic models for prediabetes, including p66shc/Shc1, to the current HbA1c-only model in terms of sensitivity, specificity and predictive accuracy. Significance was set at P ≤ 0.05. RESULTS A significant decrease of p66shc/Shc1 was determined in prediabetes compared to controls (P ≤ 0.05). HbA1c testing resulted in an accuracy of 62%, while adding p66shc and triglycerides increased predictive accuracy to 88.05%. When HbA1c was omitted and Shc1 was combined with 8-hydroxy-2'-deoxyguanosine (8-OHdG) and monocyte chemo-attractant protein-1 (MCP-1), a predictive accuracy of 89.5% was achieved. CONCLUSION Our findings showed a major improvement of sensitivity to identify prediabetes by including oxidative stress and inflammatory biomarkers underlining beneficial diagnostic information, which most likely improves prevention and early treatment options in prediabetes.
Collapse
Affiliation(s)
- Herbert F Jelinek
- Department of Biomedical EngineeringKhalifa UniversityAbu DhabiUnited Arab Emirates
- Health Engineering Innovation CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
- Biotechnology CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
| | | | - Kinda Khalaf
- Department of Biomedical EngineeringKhalifa UniversityAbu DhabiUnited Arab Emirates
- Health Engineering Innovation CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
| |
Collapse
|
12
|
Lower p66Shc promoter methylation in subjects with chronic renal failure. PLoS One 2021; 16:e0257176. [PMID: 34529688 PMCID: PMC8445414 DOI: 10.1371/journal.pone.0257176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022] Open
Abstract
Objective To determine the correlation between DNA methylation of p66Shc promoter and some markers of inflammatory and oxidative stress in chronic renal failure (CRF) patients compared with healthy subjects. Methods An observational cross-sectional study was conducted in the nephrology department at Sidi Bouzid Regional Hospital (Tunisia). In total, 39 patients with CRF and 37 healthy subjects were included. Several biochemical parameters were measured. Furthermore, markers of the oxidative and inflammatory status (MDA, TAS, SOD, and CRP) were evaluated. The p66Shc methylation status was determined using the methylation-specific PCR. Results Our results showed that levels of blood glucose, urea, creatinine, uric acid, ChT, TG, albuminuria, CRP and MDA were significantly elevated in CRF patients compared to controls. Furthermore, p66Shc promoter region was highly demethylated in CRF patients compared to healthy controls (84% vs 4%). Our data showed a positive correlation between p66Shc hypomethylation and levels of MDA (r = 0.93; p<0, 05) and CRP (r = 0.89; P <0, 05), as well as a significant negative correlation between p66Shc hypomethylation, TAS (r = -0.76; P <0, 05) and SOD (r = -0.77; p<0, 05) levels. Similarly, there was a positive correlation between p66Shc hypomethylation and the disease stages. Importantly, multiple regression analysis showed that p66shc DNA hypomethylation remains strongly correlated with MDA, CRP and stages of CRF. Conclusion This study indicates that the DNA hypomethylation of p66shc promoter was correlated with oxidative and inflammatory stress and the disease stages in CRF patients.
Collapse
|
13
|
Ren C, Zhou X, Bao X, Zhang J, Tang J, Zhu Z, Zhang N, Bai Y, Xi Y, Zhang Q, Ma B. Dioscorea zingiberensis ameliorates diabetic nephropathy by inhibiting NLRP3 inflammasome and curbing the expression of p66Shc in high-fat diet/streptozotocin-induced diabetic mice. J Pharm Pharmacol 2021; 73:1218-1229. [PMID: 34061184 DOI: 10.1093/jpp/rgab053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Diabetic nephropathy (DN) is a severe diabetic complication. Dioscorea zingiberensis (DZ) possesses excellent pharmacological properties with lower toxicity. The purpose of this study was to investigate the efficacy and mechanism of DZ in DN. METHODS DN was established by the high-fat diet combining intraperitoneal injection of streptozotocin in mice. The DZ (125 and 250 mg/kg/day) were intragastrical administered for 8 consecutive weeks. After treatment, blood, urine and kidney tissue were collected for biological detection, renal morphology, fibrosis and molecular mechanism research, respectively. KEY FINDINGS This study has shown that DZ significantly ameliorated kidney hypertrophy, renal structural damage and abnormal function of the kidney indicators (creatinine, urinary protein and blood urea nitrogen). Further molecular mechanism data suggested that the NLRP3/Cleaved-caspase-1 signal pathway was remarkably activated in DN, and DZ treatment reversed these changes, which indicated that it effectively attenuated inflammatory response caused by hyperglycaemia. In addition, DN inhibits hyperglycaemia-induced activation of oxidative stress by suppressing the expression of p66Shc proteins. CONCLUSIONS DZ could efficiently suppress oxidative stress and inflammatory responses to postpone the development of DN, and its mechanism might be related to inhibition of NLRP3 and p66Shc activities. Thus, DZ could be developed into a new therapeutic agent for DN.
Collapse
Affiliation(s)
- Chaoxing Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Xiaowei Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Xiaowen Bao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Jun Tang
- Jiangsu Huanghe Pharmaceutical Co., Ltd, Yancheng, People's Republic of China
| | - Zhiming Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Nan Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
- School of Chemical and Molecular Engineering, Nanjing Tech University, Nanjing, People's Republic of China
| | - Yu Bai
- Department of Biological Sciences, University of Toronto Scarborough, ON, Canada
| | - Youli Xi
- Department of Pharmacy, Nanjing Drum Tower Hospital, Nanjing, People's Republic of China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, People's Republic of China
| |
Collapse
|
14
|
Wang L, Tang J, Wang L, Tan F, Song H, Zhou J, Li F. Oxidative stress in oocyte aging and female reproduction. J Cell Physiol 2021; 236:7966-7983. [PMID: 34121193 DOI: 10.1002/jcp.30468] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/15/2022]
Abstract
In a healthy body, reactive oxygen species (ROS) and antioxidants remain balanced. When the balance is broken toward an overabundance of ROS, oxidative stress appears and may lead to oocyte aging. Oocyte aging is mainly reflected as the gradual decrease of oocyte quantity and quality. Here, we aim to review the relationship between oxidative stress and oocyte aging. First, we introduced that the defective mitochondria, the age-related ovarian aging, the repeated ovulation, and the high-oxygen environment were the ovarian sources of ROS in vivo and in vitro. And we also introduced other sources of ROS accumulation in ovaries, such as overweight and unhealthy lifestyles. Then, we figured that oxidative stress may act as the "initiator" for oocyte aging and reproductive pathology, which specifically causes follicular abnormally atresia, abnormal meiosis, lower fertilization rate, delayed embryonic development, and reproductive disease, including polycystic ovary syndrome and ovary endometriosis cyst. Finally, we discussed current strategies for delaying oocyte aging. We introduced three autophagy antioxidant pathways like Beclin-VPS34-Atg14, adenosine 5'-monophosphate (AMP)-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR), and p62-Keap1-Nrf2. And we also describe the different antioxidants used to combat oocyte aging. In addition, the hypoxic (5% O2 ) culture environment for oocytes avoiding oxidative stress in vitro. So, this review not only contribute to our general understanding of oxidative stress and oocyte aging but also lay the foundations for the therapies to treat premature ovarian failure and oocyte aging in women.
Collapse
Affiliation(s)
- Ling Wang
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, PR China
| | - Jinhua Tang
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, PR China
| | - Lei Wang
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, PR China
| | - Feng Tan
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, PR China
| | - Huibin Song
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, PR China
| | - Jiawei Zhou
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Fenge Li
- Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, PR China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, PR China
| |
Collapse
|
15
|
ALTamimi JZ, AlFaris NA, Al-Farga AM, Alshammari GM, BinMowyna MN, Yahya MA. Curcumin reverses diabetic nephropathy in streptozotocin-induced diabetes in rats by inhibition of PKCβ/p 66Shc axis and activation of FOXO-3a. J Nutr Biochem 2021; 87:108515. [PMID: 33017608 DOI: 10.1016/j.jnutbio.2020.108515] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/01/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023]
Abstract
This study investigated if the nephroprotective effect of Curcumin in streptozotocin-induced type 1 diabetes mellitus (DM) in rats involves downregulation/inhibition of p66Shc and examined the underlying mechanisms. Rats were divided into 4 groups (n = 12/group) as control, control + Curcumin (100 mg/kg), T1DM, and T1DM + Curcumin. Curcumin was administered orally to control or diabetic rats for 12 weeks daily. As compared to diabetic rats, Curcumin didn't affect either plasma glucose or insulin levels but significantly reduced serum levels of urea, blood urea nitrogen, and creatinine, and concurrently reduced albumin/protein urea and increased creatinine clearance. It also prevented the damage in renal tubules and mitochondria, mesangial cell expansion, the thickness of the basement membrane. Mechanistically, Curcumin reduced mRNA and protein levels of collagen I/III and transforming growth factor- β-1 (TGF-β1), reduced inflammatory cytokines levels, improved markers of mitochondrial function, and suppressed the release of cytochrome-c and the activation of caspase-3. In the kidneys of both control and diabetic rats, Curcumin reduced the levels of reactive oxygen species (ROS), increased mRNA levels of manganese superoxide dismutase (MnSOD) and gamma-glutamyl ligase, increased glutathione (GSH) and protein levels of Bcl-2 and MnSOD, and increased the nuclear levels of nuclear factor2 (Nrf2) and FOXO-3a. Besides, Curcumin reduced the nuclear activity of the nuclear factor-kappa B (NF-κB), downregulated protein kinase CβII (PKCβII), NADPH oxidase, and p66Shc, and decreased the activation of p66Shc. In conclusion, Curcumin prevents kidney damage in diabetic rats by activating Nrf2, inhibiting Nf-κB, suppressing NADPH oxidase, and downregulating/inhibiting PKCβII/p66Shc axis.
Collapse
Affiliation(s)
- Jozaa Z ALTamimi
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nora A AlFaris
- Nutrition and Food Science, Department of Physical Sport Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Ammar M Al-Farga
- Biochemistry Department, College of Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Ghedeir M Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Mohammed A Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Yang M, Han Y, Luo S, Xiong X, Zhu X, Zhao H, Jiang N, Xiao Y, Wei L, Li C, Yang J, Sun L. MAMs Protect Against Ectopic Fat Deposition and Lipid-Related Kidney Damage in DN Patients. Front Endocrinol (Lausanne) 2021; 12:609580. [PMID: 33679616 PMCID: PMC7933555 DOI: 10.3389/fendo.2021.609580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Ectopic fat deposition (EFD) in the kidney plays a key role in the development of diabetic nephropathy (DN). Mitochondria-associated ER membranes (MAMs) are structures that connect to the endoplasmic reticulum (ER) and are involved in lipid metabolism. However, there are few studies on MAMs in the field of kidney disease, and the relationship between EFD and MAMs in DN is still unclear. In this study, increased EFD in the kidneys of DN patients was observed, and analysis showed that the degree of EFD was positively correlated with renal damage. Then, the MAMs were quantified by an in situ proximity ligation assay (PLA). The MAMs in the kidneys were found to gradually decrease through the different stages of DN, while the expression of ADRP (a marker of lipid droplets) and tubulointerstitial damage increased. Moreover, correlation analysis showed that the MAMs were negatively correlated with serum lipid levels, the EFD in the kidney and renal damage. Finally, we observed decreased expression of MAM-control proteins (DsbA-L, PACS-2, and MFN-2) in different stages of DN and they were associated with lipid deposition and renal damage. These data showed that the destruction of MAMs in DN might be the cause of EFD and interstitial damage in the kidney.
Collapse
|
17
|
Hui C, Tomilov A, Garcia C, Jiang X, Fash DM, Khdour OM, Rosso C, Filippini G, Prato M, Graham J, Hecht S, Havel P, Cortopassi G. Novel idebenone analogs block Shc's access to insulin receptor to improve insulin sensitivity. Biomed Pharmacother 2020; 132:110823. [PMID: 33045613 DOI: 10.1016/j.biopha.2020.110823] [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: 07/19/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 10/23/2022] Open
Abstract
There has been little innovation in identifying novel insulin sensitizers. Metformin, developed in the 1920s, is still used first for most Type 2 diabetes patients. Mice with genetic reduction of p52Shc protein have improved insulin sensitivity and glucose tolerance. By high-throughput screening, idebenone was isolated as the first small molecule 'Shc Blocker'. Idebenone blocks p52Shc's access to Insulin Receptor to increase insulin sensitivity. In this work the avidity of 34 novel idebenone analogs and 3 metabolites to bind p52Shc, and to block the interaction of p52Shc with the Insulin receptor was tested. Our hypothesis was that if an idebenone analog bound and blocked p52Shc's access to insulin receptor better than idebenone, it should be a more effective insulin sensitizing agent than idebenone itself. Of 34 analogs tested, only 2 both bound p52Shc more tightly and/or blocked the p52Shc-Insulin Receptor interaction more effectively than idebenone. Of those 2 only idebenone analog #11 was a superior insulin sensitizer to idebenone. Also, the long-lasting insulin-sensitizing potency of idebenone in rodents over many hours had been puzzling, as the parent molecule degrades to metabolites within 1 h. We observed that two of the idebenone's three metabolites are insulin sensitizing almost as potently as idebenone itself, explaining the persistent insulin sensitization of this rapidly metabolized molecule. These results help to identify key SAR = structure-activity relationship requirements for more potent small molecule Shc inhibitors as Shc-targeted insulin sensitizers for type 2 diabetes.
Collapse
Affiliation(s)
- ChunKiu Hui
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Alexey Tomilov
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Chase Garcia
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - XiaoSong Jiang
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - David M Fash
- Center for BioEnergetics, Biodesign Institute, Arizona State University, 1001 S McAllister Ave, Tempe, AZ, 85287, USA.
| | - Omar M Khdour
- Center for BioEnergetics, Biodesign Institute, Arizona State University, 1001 S McAllister Ave, Tempe, AZ, 85287, USA.
| | - Cristian Rosso
- Department of Chemical and Pharmaceutical Sciences, CENMAT, Center of Excellence for Nanostructured Materials, INSTM UdR, Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy.
| | - Giacomo Filippini
- Department of Chemical and Pharmaceutical Sciences, CENMAT, Center of Excellence for Nanostructured Materials, INSTM UdR, Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy.
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, CENMAT, Center of Excellence for Nanostructured Materials, INSTM UdR, Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy; Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014, Donostia San Sebastián, Spain; Basque Fdn Sci, Ikerbasque, Bilbao, 48013, Spain.
| | - James Graham
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Sidney Hecht
- Center for BioEnergetics, Biodesign Institute, Arizona State University, 1001 S McAllister Ave, Tempe, AZ, 85287, USA; School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA.
| | - Peter Havel
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Gino Cortopassi
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| |
Collapse
|
18
|
Functional Role of p53 in the Regulation of Chemical-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6039769. [PMID: 32190175 PMCID: PMC7066401 DOI: 10.1155/2020/6039769] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
The nuclear transcription factor p53, discovered in 1979, has a broad range of biological functions, primarily the regulation of apoptosis, the cell cycle, and DNA repair. In addition to these canonical functions, a growing body of evidence suggests that p53 plays an important role in regulating intracellular redox homeostasis through transcriptional and nontranscriptional mechanisms. Oxidative stress induction and p53 activation are common responses to chemical exposure and are suggested to play critical roles in chemical-induced toxicity. The activation of p53 can exert either prooxidant or antioxidant activity, depending on the context. In this review, we discuss the functional role of p53 in regulating chemical-induced oxidative stress, summarize the potential signaling pathways involved in p53's regulation of chemically mediated oxidative stress, and propose issues that should be addressed in future studies to improve understanding of the relationship between p53 and chemical-induced oxidative stress.
Collapse
|
19
|
Zhu H, Liu X, Zhang C, Li Q, An X, Liu S, Wu L, Zhang B, Yuan Y, Xing C. Association of urinary acidification function with the progression of diabetic kidney disease in patients with type 2 diabetes. J Diabetes Complications 2019; 33:107419. [PMID: 31473080 DOI: 10.1016/j.jdiacomp.2019.107419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/10/2019] [Accepted: 08/16/2019] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Although diabetic kidney disease (DKD) has been considered as a glomerulocentric disease in the past few decades, growing evidence demonstrated that tubular damage was indispensable in its pathogenesis and progression. This study was designed to investigate the association of urinary acidification dysfunction with the progression of DKD in type 2 diabetic patients. METHODS Here the urinary acidification functions were measured from 80 participants with renal biopsy-proven DKD. The different kinds of renal tubular transportation dysfunction were analyzed, including the dysfunction of bicarbonate reabsorption, titratable acid secretion, and ammonium secretion. In addition, patients were followed up for 17 (interquartile range, 11-32) months to evaluate the effect of urinary acidification dysfunction in the progression of DKD. RESULTS The most common urinary acidification dysfunction was the disorder of ammonium secretion, accounting for 53.75%. The more proteinuria excretion and the lower glomerular filtration rate (GFR) were observed in the urinary titratable acid disorder group than the normal group, and the same results were obtained for ammonium secretion disorder. Urine titratable acid was positively correlated with eGFR whereas it was inversely correlated with proteinuria, serum creatinine, and BUN. Moreover, 24 h urine protein, serum creatinine, BUN and cystatin C increased from DKD stage II to stage IV, whereas the eGFR and urine titratable acid decreased in the same way. Furthermore, Kaplan-Meier analysis and Cox regression showed that the disorder of titratable acid was an independent risk factor for DKD progression. CONCLUSIONS The dysfunction of urinary titratable acid is a potential biomarker for the severity of proteinuria, eGFR and glomerular lesions in patients with DKD. Moreover, the titratable acid disorder is an independent risk factor of the DKD progression.
Collapse
MESH Headings
- Acidosis, Renal Tubular/complications
- Acidosis, Renal Tubular/diagnosis
- Acidosis, Renal Tubular/epidemiology
- Acidosis, Renal Tubular/etiology
- Acids/analysis
- Acids/metabolism
- Adult
- Aged
- Biomarkers/metabolism
- Biomarkers/urine
- Comorbidity
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/urine
- Diabetic Nephropathies/complications
- Diabetic Nephropathies/epidemiology
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/urine
- Disease Progression
- Female
- Glomerular Filtration Rate
- Humans
- Hydrogen-Ion Concentration
- Kidney/metabolism
- Kidney/physiopathology
- Kidney Failure, Chronic/diagnosis
- Kidney Failure, Chronic/epidemiology
- Kidney Failure, Chronic/physiopathology
- Kidney Failure, Chronic/urine
- Male
- Middle Aged
- Prognosis
- Proteinuria/epidemiology
- Proteinuria/etiology
- Proteinuria/urine
- Urine/chemistry
Collapse
Affiliation(s)
- Huanhuan Zhu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Xi Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Chengning Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Qing Li
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Xiaofei An
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Simeng Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Lin Wu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Bo Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yanggang Yuan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| | - Changying Xing
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
20
|
Zheng D, Tao M, Liang X, Li Y, Jin J, He Q. p66Shc regulates podocyte autophagy in high glucose environment through the Notch-PTEN-PI3K/Akt/mTOR pathway. Histol Histopathol 2019; 35:405-415. [PMID: 31650524 DOI: 10.14670/hh-18-178] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Autophagy has been found to be involved in podocyte injury, which is a key factor in the progression of diabetic kidney disease (DKD). p66Shc is an important protein adaptor that regulates production of reactive oxygen species (ROS) and induction of apoptosis, and is a novel biomarker for oxidative damage of renal tubules. Our preliminary studies showed that p66Shc expression in podocytes of DKD patients is increased, while autophagic flux and podocyte number is decreased in DKD patients. The mechanism by which p66Shc may regulate podocyte autophagy and injury remains unknown. The present study aimed to investigate the molecular function of p66Shc under high glucose condition and its possible therapeutic utility in DKD. METHODS We histologically evaluated kidney injury in a streptozocin (STZ)-induced mouse model of diabetes using HE, PAS, PASM, and Masson staining and assessed glomerular structure by transmission electron microscopy. The apoptosis rate of high glucose-treated podocytes was assessed by TUNEL and Annexin V/PI staining. Markers of podocyte autophagy were measured by immunofluorescence and western blotting. DHE/ET fluorescence quantification was used for ROS detection and quantification. RESULTS Urine creatinine, serum creatinine, urinary microalbumin, and p66Shc expression were significantly increased in STZ-induced diabetic mice. Cultured MPC5 podocytes subjected to high glucose showed reduced viability, and p66Shc overexpression further accelerated apoptosis. p66Shc knockdown enhanced HG-induced autophagy, while p66Shc overexpression reduced the expression of PTEN and increased the expression of mTOR and phospho-mTOR. LC3 protein expression was higher in cells with p66Shc knockdown, indicating that activation of p66Shc inhibits podocyte autophagy. DAPT, an inhibitor of the Notch pathway, downregulated the expression of p66Shc. CONCLUSION These findings indicate that p66Shc inhibits podocyte autophagy and induces apoptosis through the Notch -PTEN-PI3K/Akt/ mTOR signaling pathway in high glucose environment, providing novel evidence for its potential role in DKD treatment.
Collapse
Affiliation(s)
- Danna Zheng
- Zhejiang Chinese Medical University, Zhejiang, PR China.,Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, PR China.,People's Hospital of Hangzhou Medical College, Zhejiang, PR China.,Chinese Medical Nephrology Key Laboratory of Zhejiang Province, Zhejiang, PR China
| | - Mei Tao
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, PR China.,People's Hospital of Hangzhou Medical College, Zhejiang, PR China.,Chinese Medical Nephrology Key Laboratory of Zhejiang Province, Zhejiang, PR China
| | - Xudong Liang
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, PR China.,People's Hospital of Hangzhou Medical College, Zhejiang, PR China.,Chinese Medical Nephrology Key Laboratory of Zhejiang Province, Zhejiang, PR China
| | - Yiwen Li
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, PR China.,People's Hospital of Hangzhou Medical College, Zhejiang, PR China.,Chinese Medical Nephrology Key Laboratory of Zhejiang Province, Zhejiang, PR China
| | - Juan Jin
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, PR China.,People's Hospital of Hangzhou Medical College, Zhejiang, PR China.,Chinese Medical Nephrology Key Laboratory of Zhejiang Province, Zhejiang, PR China
| | - Qiang He
- Department of Nephrology, Zhejiang Provincial People's Hospital, Zhejiang, PR China.,People's Hospital of Hangzhou Medical College, Zhejiang, PR China.,Chinese Medical Nephrology Key Laboratory of Zhejiang Province, Zhejiang, PR China
| |
Collapse
|
21
|
Mishra M, Duraisamy AJ, Bhattacharjee S, Kowluru RA. Adaptor Protein p66Shc: A Link Between Cytosolic and Mitochondrial Dysfunction in the Development of Diabetic Retinopathy. Antioxid Redox Signal 2019; 30:1621-1634. [PMID: 30105917 PMCID: PMC6459280 DOI: 10.1089/ars.2018.7542] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Diabetes increases oxidative stress in the retina and dysfunctions their mitochondria, accelerating capillary cell apoptosis. A 66 kDa adaptor protein, p66Shc, is considered as a sensor of oxidative stress-induced apoptosis. In the pathogenesis of diabetic retinopathy, a progressive disease, reactive oxygen species (ROS) production by activation of a small molecular weight G-protein (Ras-related C3 botulinum toxin substrate 1 [Rac1])-Nox2 signaling precedes mitochondrial damage. Rac1 activation is facilitated by guanine exchange factors (GEFs), and p66Shc increases Rac1-specific GEF activity of Son of Sevenless 1 (Sos1). p66Shc also possesses oxidoreductase activity and can directly stimulate mitochondrial ROS generation. Our aim was to investigate the role of p66Shc in the development of diabetic retinopathy and mechanism of its transcription. RESULTS High glucose increased p66Shc expression in human retinal endothelial cells, and elevated acetylated histone 3 lysine 9 (H3K9) levels and transcriptional factor p53 binding at its promoter. Glucose also augmented interactions between Rac1 and Sos1 and activated Rac1-Nox2. Phosphorylation of p66Shc was increased, allowing it to interact with peptidyl prolyl isomerase to facilitate its localization inside the mitochondria, culminating in mitochondrial damage. P66shc-small interfering RNA (siRNA) inhibited glucose-induced Rac1 activation and mitochondrial damage. Similar results are observed in retinal microvessels from diabetic rats. INNOVATION This is the first report identifying the role of p66Shc in the development of diabetic retinopathy and implicating increased histone acetylation in its transcriptional regulation. CONCLUSION Thus, p66Shc has dual role in the development of diabetic retinopathy; its regulation in the early stages of the disease should impede Rac1-ROS production and, in the later stages, prevent mitochondrial damage and initiation of a futile cycle of free radicals.
Collapse
Affiliation(s)
- Manish Mishra
- 1 Department of Ophthalmology, Kresge Eye Institute, Wayne State University, Detroit, Michigan
| | - Arul J Duraisamy
- 1 Department of Ophthalmology, Kresge Eye Institute, Wayne State University, Detroit, Michigan
| | - Sudarshan Bhattacharjee
- 1 Department of Ophthalmology, Kresge Eye Institute, Wayne State University, Detroit, Michigan
| | - Renu A Kowluru
- 1 Department of Ophthalmology, Kresge Eye Institute, Wayne State University, Detroit, Michigan.,2 Department of Anatomy/Cell Biology, Kresge Eye Institute, Wayne State University, Detroit, Michigan
| |
Collapse
|
22
|
Wang W, Sun W, Cheng Y, Xu Z, Cai L. Role of sirtuin-1 in diabetic nephropathy. J Mol Med (Berl) 2019; 97:291-309. [PMID: 30707256 PMCID: PMC6394539 DOI: 10.1007/s00109-019-01743-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/29/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Diabetic nephropathy (DN) is a research priority for scientists around the world because of its high prevalence and poor prognosis. Although several mechanisms have been shown to be involved in its pathogenesis and many useful drugs have been developed, the management of DN remains challenging. Increasing amounts of evidence show that silent information regulator 2 homolog 1 (sirtuin-1), a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, plays a crucial role in the pathogenesis and development of DN. Clinical data show that gene polymorphisms of sirtuin-1 affect patient vulnerability to DN. In addition, upregulation of sirtuin-1 attenuates DN in various experimental models of diabetes and in renal cells, including podocytes, mesangial cells, and renal proximal tubular cells, incubated with high concentrations of glucose or advanced glycation end products. Mechanistically, sirtuin-1 has its renoprotective effects by modulating metabolic homeostasis and autophagy, resisting apoptosis and oxidative stress, and inhibiting inflammation through deacetylation of histones and the transcription factors p53, forkhead box group O, nuclear factor-κB, hypoxia-inducible factor-1α, and others. Furthermore, some microRNAs have been implicated in the progression of DN because they target sirtuin-1 mRNA. Several synthetic drugs and natural compounds have been identified that upregulate the expression and activity of sirtuin-1, which protects against DN. The present review will summarize advances in knowledge regarding the role of sirtuin-1 in the pathogenesis of DN. The available evidence implies that sirtuin-1 has great potential as a clinical target for the prevention and treatment of diabetes.
Collapse
Affiliation(s)
- Wanning Wang
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
- Pediatric Research Institute, Department of Pediatrics, The University of Louisville School of Medicine, Louisville, KY 40292 USA
| | - Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
| | - Yanli Cheng
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin Province China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, The University of Louisville School of Medicine, Louisville, KY 40292 USA
- Departments of Radiation Oncology, Pharmacology and Toxicology, The University of Louisville School of Medicine, 570 S. Preston Str., Baxter I, Suite 304F, Louisville, KY 40292 USA
| |
Collapse
|
23
|
Chen X, Han Y, Gao P, Yang M, Xiao L, Xiong X, Zhao H, Tang C, Chen G, Zhu X, Yuan S, Liu F, Dong LQ, Liu F, Kanwar YS, Sun L. Disulfide-bond A oxidoreductase-like protein protects against ectopic fat deposition and lipid-related kidney damage in diabetic nephropathy. Kidney Int 2019; 95:880-895. [PMID: 30791996 DOI: 10.1016/j.kint.2018.10.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 12/31/2022]
Abstract
Ectopic fat deposition (EFD) in the kidney has been shown to play a causal role in diabetic nephropathy; however, the mechanism underlying EFD remains elusive. By transcriptome analysis, we found decreased expression levels of disulfide-bond A oxidoreductase-like protein (DsbA-L) in the kidneys of diabetic mice (induced by high-fat diet plus Streptozotocin) compared with control mice. Increased expression of adipocyte differentiation-related protein and abnormal levels of collagen I, fibronectin, and phosphorylated 5'AMP-activated kinase (p-AMPK), adipose triglyceride lipase (p-ATGL), and HMG-CoA reductase (p-HMGCR) were also observed in diabetic mice. These alterations were accompanied by deposition of lipid droplets in the kidney, and were more pronounced in diabetic DsbA-L knockout mice. In vitro, overexpression of DsbA-L ameliorated high glucose-induced intracellular lipid droplet deposition in a human proximal tubular cell line, and DsbA-L siRNA aggravated lipid droplet deposition and reduced the levels of p-AMPK, p-ATGL, and p-HMGCR. High glucose and palmitic acid treatment enhanced the expression of interleukin-1β and interleukin-18; these enhancements were further increased after treatment with DsbA-L siRNA but alleviated by co-treatment with an AMPK activator. In kidney biopsy tissue from patients with diabetic nephropathy, DsbA-L expression was negatively correlated with EFD and tubular damage. Collectively, these results suggest that DsbA-L has a protective role against EFD and lipid-related kidney damage in diabetic nephropathy. Activation of the AMPK pathway is a potential mechanism underlying DsbA-L action in the kidney.
Collapse
Affiliation(s)
- Xianghui Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Gao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofen Xiong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuejing Zhu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuguang Yuan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lily Q Dong
- Department of Cell Systems & Anatomy, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Feng Liu
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Yashpal S Kanwar
- Departments of Pathology & Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Ectopic lipid accumulation: potential role in tubular injury and inflammation in diabetic kidney disease. Clin Sci (Lond) 2018; 132:2407-2422. [PMID: 30348828 DOI: 10.1042/cs20180702] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/18/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022]
Abstract
Emerging studies suggest that lipid accumulates in the kidneys during diabetic kidney disease (DKD). However, the correlation between ectopic lipid accumulation with tubular damage has not been thoroughly elucidated to date. Using Oil Red staining, lipid accumulation was observed in the kidneys of type 2 DKD patients (classes II-III) and db/db mice compared with the control and was predominantly located in the proximal tubular compartment. Immunohistochemistry (IHC) staining showed that the intensity of adipose differentiation related protein (ADRP) and sterol regulatory element binding protein-1 (SREBP-1) was clearly up-regulated, which was positively correlated with the tubulointerstitial damage score and inflammation. Furthermore, the urine ADRP content significantly increased in DKD patients compared with the control, which positively correlated with abnormal lipid metabolism, serum creatinine, urine N-acetyl-β-glucosaminidase (NAG), albumin excretion (albumin-to-creatinine ratio (ACR)), and tumor necrosis factor-α (TNF-α) expression. However, there was no significant difference observed in plasma ADRP levels. In addition, the expression of SREBP-1 protein was dramatically increased in peripheral blood mononuclear cells (PBMCs) isolated from DKD patients, which was also tightly correlated with urine NAG, ACR, and TNF-α levels. In vitro studies demonstrated increased ADRP and SREBP-1 expression accompanied by lipid accumulation in HK-2 cells cultured in high glucose (HG). HG induced high levels of TNF-α expression, which was partially blocked by transfection of ADRP siRNA or SREBP-1 siRNA. These data indicated that ADRP and SREBP-1 are crucial factors that mediate lipid accumulation with tubular damage and inflammation in DKD, and ectopic lipid accumulation may serve as a novel therapeutic target for amelioration of tubular injury in DKD.
Collapse
|
25
|
Miller BS, Blumenthal SR, Shalygin A, Wright KD, Staruschenko A, Imig JD, Sorokin A. Inactivation of p66Shc Decreases Afferent Arteriolar K ATP Channel Activity and Decreases Renal Damage in Diabetic Dahl SS Rats. Diabetes 2018; 67:2206-2212. [PMID: 30131395 PMCID: PMC6198347 DOI: 10.2337/db18-0308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/16/2018] [Indexed: 12/17/2022]
Abstract
Increased expression of adaptor protein p66Shc has been associated with progression of diabetic nephropathy. Afferent arteriolar dilation and glomerular hyperfiltration in diabetes are due to increased KATP channel availability and activity. Hyperglycemia was induced in Dahl salt-sensitive (SS) rats in a model of diabetes induced by streptozotocin (STZ). Renal injury was evaluated in SS rats and genetically modified SS rats either lacking p66Shc (p66Shc knockout [p66ShcKO]) or expressing p66Shc mutant (p66Shc-S36A). Afferent arteriolar diameter responses during STZ-induced hyperfiltration were determined by using the juxtamedullary nephron technique. Albuminuria and glomerular injury were mitigated in p66ShcKO and p66Shc-S36A rats with STZ-induced diabetes. SS rats with STZ-induced diabetes had significantly increased afferent arteriolar diameter, whereas p66ShcKO and p66Shc-S36A rats did not. SS rats with STZ-induced diabetes, but not p66ShcKO or p66Shc-S36A rats with STZ-induced diabetes, had an increased vasodilator response to the KATP channel activator pinacidil. Likewise, the KATP inhibitor glibenclamide resulted in a greater decrease in afferent arteriolar diameter in SS rats with STZ-induced diabetes than in STZ-treated SS p66ShcKO and p66Shc-S36A rats. Using patch-clamp electrophysiology, we demonstrated that p66ShcKO decreases KATP channel activity. These results indicate that inactivation of the adaptor protein p66Shc decreases afferent arteriolar KATP channel activity and decreases renal damage in diabetic SS rats.
Collapse
Affiliation(s)
- Bradley S Miller
- Department of Medicine, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
| | - Shoshana R Blumenthal
- Department of Medicine, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI
| | - Alexey Shalygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Kevin D Wright
- Department of Medicine, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
| | - Alexander Staruschenko
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - John D Imig
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Andrey Sorokin
- Department of Medicine, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
26
|
Cao W, Liu X, Xu X, Zeng M, Sun B, Yu X, Wang N, Mao H, Zhang B, Yuan Y, Xing C. The Src homology and collagen A (ShcA) adaptor protein may participate in the pathogenesis of membranous lupus nephritis. Lupus 2018; 27:2014-2019. [PMID: 30189773 DOI: 10.1177/0961203318796295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The Src homology and collagen A (ShcA) adaptor protein that binds to tyrosine kinase receptors. ShcA plays a role in insulin signaling, stress resistance and energy metabolism. The 66-kDa Src homology 2 domain-containing protein (p66shc) belongs to the ShcA family and has been associated with reactive oxygen species (ROS); increased ROS is involved in the pathology of lupus nephritis (LN). However, whether ShcA can act as a biomarker for oxidative injury in LN is unknown. This study is aimed to investigate the ShcA expression in kidney tissues from patients presenting with LN and the association between ShcA expression and clinical parameters. Renal biopsy tissues were obtained from 62 LN, 20 primary membranous nephropathy (MN) and 10 other secondary MN patients. ShcA was measured by immunofluorescence. The expression of ShcA in the membranous lupus nephritis (class V) group showed a higher trend but there were no significant differences compared with pure mesangial disease (class II) and proliferative (Class III/IV) lupus nephritis. ShcA deposits were negative in primary and other secondary MN. ShcA might act as a new biomarker and a diagnostic tool to identify membranous lupus nephritis with other MN.
Collapse
Affiliation(s)
- W Cao
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - X Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - X Xu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - M Zeng
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - B Sun
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - X Yu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - N Wang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - H Mao
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - B Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Y Yuan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - C Xing
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Perturbations in mitochondrial dynamics by p66Shc lead to renal tubular oxidative injury in human diabetic nephropathy. Clin Sci (Lond) 2018; 132:1297-1314. [PMID: 29760122 DOI: 10.1042/cs20180005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 12/26/2022]
Abstract
Renal tubular injury is increasingly being recognized as an early characteristic of diabetic nephropathy (DN). Mitochondrial dynamic alterations and redox protein p66Shc-mediated oxidative stress are both critical for ensuing diabetic tubular cell injury and apoptosis; whether these two processes are interlinked remains unclear. In the present study, we observed changes in mitochondrial morphology and expression of associated proteins in tubules of patients with DN. We demonstrated mitochondrial fragmentation as an important pathogenic feature of tubular cell injury that is linked to oxidative stress and p66Shc up-regulation. In renal proximal tubular cells, alterations in mitochondrial dynamics and expression of fission-fusion proteins were observed under high glucose (HG) ambience, along with p66Shc Ser36 phosphorylation. Gene ablation of p66Shc alleviated HG-induced mitochondrial fragmentation, down-regulated Fis1 and reduced p66Shc-Fis1 binding, increased Mfn1 expression, and disrupted interactions between Mfn1 and proapoptotic Bak. Overexpression of p66Shc exacerbated these changes, whereas overexpression of dominant-negative p66Shc Ser36 mutant had a marginal effect under HG, indicating that p66Shc phosphorylation as a prerequisite in the modulation of mitochondrial dynamics. Disrupted mitochondrial dynamics and enhanced Mfn1-Bak interactions modulated by p66Shc led to loss of mitochondrial voltage potential, cytochrome C release, excessive ROS generation, and apoptosis. Taken together, these results link p66Shc to mitochondrial dynamic alterations in the pathogenesis of DN and unveil a novel mechanism by which p66Shc mediates HG-induced mitochondrial fragmentation and proapoptotic signaling that results in oxidative injury and apoptosis in the tubular compartment in human diabetic nephropathy.
Collapse
|
28
|
Fadini GP, Albiero M, Bonora BM, Poncina N, Vigili de Kreutzenberg S, Avogaro A. p66Shc gene expression in peripheral blood mononuclear cells and progression of diabetic complications. Cardiovasc Diabetol 2018; 17:16. [PMID: 29343271 PMCID: PMC5771224 DOI: 10.1186/s12933-018-0660-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 01/06/2018] [Indexed: 12/26/2022] Open
Abstract
Background The risk of diabetic complications is modified by genetic and epigenetic factors. p66Shc drives the hyperglycaemic cell damage and its deletion prevents experimental diabetic complications. We herein tested whether p66Shc expression in peripheral blood mononuclear cells (PBMCs) predicts adverse outcomes in people with diabetes. Methods In a cohort of 100 patients with diabetes (16 type 1 and 84 type 2), we quantified baseline p66Shc expression in PBMCs by quantitative PCR. Patients were extensively characterized for demographics, anthropometrics, biochemical data, prevalence of complications, and medications. With a pseudo-prospective design, we retrieved cardiovascular death, major adverse cardiovascular events (MACE), and new occurrence of micro- or macroangiopathy during follow-up. Results At baseline, patients were on average 60 year old, with 10-year diabetes duration, and overall poor glycaemic control (HbA1c 7.8%). Patients with high versus low p66Shc expression (based on median value) had very similar baseline characteristics. Average p66Shc expression did not differ by presence/absence of complications. During a median 5.6-year follow-up, the primary endpoint of cardiovascular death or MACE occurred in 22 patients, but no relation was detected between cardiovascular outcomes and p66Shc expression. In patients who developed new complications at follow-up, baseline p66Shc was significantly higher, especially for macroangiopathy. The incidence of new macroangiopathy was > 3-times higher in patients with high versus those with low baseline p66Shc expression. Conclusions p66Shc expression in PBMCs was not associated with prevalent diabetic complications but predicted new onset of complications, especially macroangiopathy, although no relation with hard cardiovascular endpoints was detected.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35100, Padua, Italy. .,Venetian Institute of Molecular Medicine, 35100, Padua, Italy.
| | - Mattia Albiero
- Venetian Institute of Molecular Medicine, 35100, Padua, Italy
| | | | - Nicol Poncina
- Venetian Institute of Molecular Medicine, 35100, Padua, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Via Giustiniani 2, 35100, Padua, Italy
| |
Collapse
|
29
|
Boengler K, Bencsik P, Palóczi J, Kiss K, Pipicz M, Pipis J, Ferdinandy P, Schlüter KD, Schulz R. Lack of Contribution of p66shc and Its Mitochondrial Translocation to Ischemia-Reperfusion Injury and Cardioprotection by Ischemic Preconditioning. Front Physiol 2017; 8:733. [PMID: 29051737 PMCID: PMC5633811 DOI: 10.3389/fphys.2017.00733] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/08/2017] [Indexed: 12/23/2022] Open
Abstract
Whereas high amounts of reactive oxygen species (ROS) contribute to cardiac damage following ischemia and reperfusion (IR), low amounts function as trigger molecules in the cardioprotection by ischemic preconditioning (IPC). The mitochondrial translocation and contribution of the hydrogen peroxide-generating protein p66shc in the cardioprotection by IPC is unclear yet. In the present study, we investigated the mitochondrial translocation of p66shc, addressed the impact of p66shc on ROS formation after IR, and characterized the role of p66shc in IR injury per se and in the cardioprotection by IPC. The amount of p66shc in subsarcolemmal (SSM) and interfibrillar mitochondria (IFM) isolated from wildtype mouse left ventricles (LV) was determined after 40 min normoxic perfusion and after 30 min ischemia and 10 min reperfusion without and with IPC. The p66shc content in SSM (in % of normoxic controls, n = 5) was 174 ± 16% (n = 6, p < 0.05) after IR, and was reduced to 128 ± 13% after IPC (n = 6, p = ns). In IFM, the amount of p66shc remained unchanged (IR: 81 ± 7%, n = 6; IPC: 110 ± 5%, n = 6, p = ns). IR induced an increase in ROS formation in SSM and IFM isolated from mouse wildtype LV, which was more pronounced in SSM than in IFM (1.18 ± 0.18 vs. 0.81 ± 0.16, n = 6, p < 0.05). In mitochondria from p66shc-knockout mice (p66shc-KO), the increase in ROS formation by IR was not different between SSM and IFM (0.90 ± 0.11 vs. 0.73 ± 0.08, n = 6, p = ns). Infarct size (in % of the left ventricle) was 51.7 ± 2.9% in wildtype and 59.7 ± 3.8% in p66shc-KO hearts in vitro and was significantly reduced to 35.8 ± 4.4% (wildtype) and 34.7 ± 5.6% (p66shc-KO) by IPC, respectively. In vivo, infarct size was 57.8 ± 2.9% following IR (n = 9) and was reduced to 40.3 ± 3.5% by IPC (n = 11, p < 0.05) in wildtype mice. In p66shc-knockout mice, infarct sizes were similar to those measured in wildtype animals (IR: 56.2 ± 4.3%, n = 11; IPC: 42.1 ± 3.9%, n = 13, p < 0.05). Taken together, the mitochondrial translocation of p66shc following IR and IPC differs between mitochondrial populations. However, similar infarct sizes after IR and preserved infarct size reductions by IPC in p66shc-KO mice suggest that p66shc-derived ROS are not involved in the cardioprotection by IPC nor do they contribute to IR injury per se.
Collapse
Affiliation(s)
- Kerstin Boengler
- Physiologisches Institut, Justus-Liebig-Universität, Giessen, Germany
| | - Péter Bencsik
- Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - János Palóczi
- Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Krisztina Kiss
- Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Márton Pipicz
- Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| | | | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | | | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität, Giessen, Germany
| |
Collapse
|
30
|
Wright KD, Staruschenko A, Sorokin A. Role of adaptor protein p66Shc in renal pathologies. Am J Physiol Renal Physiol 2017; 314:F143-F153. [PMID: 28978535 DOI: 10.1152/ajprenal.00414.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
p66Shc is one of the three adaptor proteins encoded by the Shc1 gene, which are expressed in many organs, including the kidney. Recent studies shed new light on several key questions concerning the signaling mechanisms mediated by p66Shc. The central goal of this review article is to summarize recent findings on p66Shc and the role it plays in kidney physiology and pathology. This article provides a review of the various mechanisms whereby p66Shc has been shown to function within the kidney through a wide range of actions. The mitochondrial and cytoplasmic signaling of p66Shc, as it relates to production of reactive oxygen species (ROS) and renal pathologies, is further discussed.
Collapse
Affiliation(s)
- Kevin D Wright
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Alexander Staruschenko
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Andrey Sorokin
- Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
31
|
Papadopoulou-Marketou N, Kanaka-Gantenbein C, Marketos N, Chrousos GP, Papassotiriou I. Biomarkers of diabetic nephropathy: A 2017 update. Crit Rev Clin Lab Sci 2017; 54:326-342. [DOI: 10.1080/10408363.2017.1377682] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nektaria Papadopoulou-Marketou
- Diabetes Centre of the Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Athens, Greece
- Department of Endocrinology, Department of Medical and Health Sciences, Linkoping University, Linkoping, Sweden
| | - Christina Kanaka-Gantenbein
- Diabetes Centre of the Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Athens, Greece
| | | | - George P. Chrousos
- Diabetes Centre of the Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, “Aghia Sophia” Children’s Hospital, Athens, Greece
| |
Collapse
|
32
|
Krochmal M, Cisek K, Filip S, Markoska K, Orange C, Zoidakis J, Gakiopoulou C, Spasovski G, Mischak H, Delles C, Vlahou A, Jankowski J. Identification of novel molecular signatures of IgA nephropathy through an integrative -omics analysis. Sci Rep 2017; 7:9091. [PMID: 28831120 PMCID: PMC5567309 DOI: 10.1038/s41598-017-09393-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022] Open
Abstract
IgA nephropathy (IgAN) is the most prevalent among primary glomerular diseases worldwide. Although our understanding of IgAN has advanced significantly, its underlying biology and potential drug targets are still unexplored. We investigated a combinatorial approach for the analysis of IgAN-relevant -omics data, aiming at identification of novel molecular signatures of the disease. Nine published urinary proteomics datasets were collected and the reported differentially expressed proteins in IgAN vs. healthy controls were integrated into known biological pathways. Proteins participating in these pathways were subjected to multi-step assessment, including investigation of IgAN transcriptomics datasets (Nephroseq database), their reported protein-protein interactions (STRING database), kidney tissue expression (Human Protein Atlas) and literature mining. Through this process, from an initial dataset of 232 proteins significantly associated with IgAN, 20 pathways were predicted, yielding 657 proteins for further analysis. Step-wise evaluation highlighted 20 proteins of possibly high relevance to IgAN and/or kidney disease. Experimental validation of 3 predicted relevant proteins, adenylyl cyclase-associated protein 1 (CAP1), SHC-transforming protein 1 (SHC1) and prolylcarboxypeptidase (PRCP) was performed by immunostaining of human kidney sections. Collectively, this study presents an integrative procedure for -omics data exploitation, giving rise to biologically relevant results.
Collapse
Affiliation(s)
- Magdalena Krochmal
- Biomedical Research Foundation Academy of Athens, Center of Basic Research, Athens, Greece
- RWTH Aachen University Hospital, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | | | - Szymon Filip
- Biomedical Research Foundation Academy of Athens, Center of Basic Research, Athens, Greece
| | - Katerina Markoska
- Department of Nephrology, Medical Faculty, University of Skopje, Skopje, Macedonia
| | - Clare Orange
- Department of Pathology, School of Medicine, University of Glasgow, Glasgow, UK
| | - Jerome Zoidakis
- Biomedical Research Foundation Academy of Athens, Center of Basic Research, Athens, Greece
| | - Chara Gakiopoulou
- Pathology Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Goce Spasovski
- Department of Nephrology, Medical Faculty, University of Skopje, Skopje, Macedonia
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, Glasgow, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Antonia Vlahou
- Biomedical Research Foundation Academy of Athens, Center of Basic Research, Athens, Greece.
| | - Joachim Jankowski
- RWTH Aachen University Hospital, Institute for Molecular Cardiovascular Research, Aachen, Germany.
- University of Maastricht, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands.
| |
Collapse
|
33
|
Yang S, Zhao L, Han Y, Liu Y, Chen C, Zhan M, Xiong X, Zhu X, Xiao L, Hu C, Liu F, Zhou Z, Kanwar YS, Sun L. Probucol ameliorates renal injury in diabetic nephropathy by inhibiting the expression of the redox enzyme p66Shc. Redox Biol 2017; 13:482-497. [PMID: 28728079 PMCID: PMC5514499 DOI: 10.1016/j.redox.2017.07.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 12/19/2022] Open
Abstract
AIMS Probucol is an anti-hyperlipidemic agent and a potent antioxidant drug that can delay progression of diabetic nephropathy (DN) and reverses renal oxidative stress in diabetic animal models; however, the mechanisms underlying these effects remain unclear. p66Shc is a newly recognized mediator of mitochondrial ROS production in renal cells under high-glucose (HG) ambience. We previously showed that p66Shc can serve as a biomarker for renal oxidative injury in DN patients and that p66Shc up-regulation is correlated with renal damage in vivo and in vitro. Here, we determined whether probucol ameliorates renal injury in DN by inhibiting p66Shc expression. RESULTS We found that the expression of SIRT1, Ac-H3 and p66Shc in kidneys of DN patients was altered. Also, probucol reduced the levels of serum creatinine, urine protein and LDL-c and attenuated renal oxidative injury and fibrosis in STZ induced diabetic mice. In addition, probucol reversed p-AMPK, SIRT1, Ac-H3 and p66Shc expression. Correlation analyses showed that p66Shc expression was correlated with p-AMPK and Sirt1 expression and severity of renal injury. In vitro pretreatment of HK-2 cells with p-AMPK and SIRT1 siRNA negated the beneficial effects of probucol. Furthermore, we noted that probucol activates p-AMPK and Sirt1 and inhibits p66shc mRNA transcription by facilitating the binding of Sirt1 to the p66Shc promoter and modulation of Ac-H3 expression in HK-2 cells under HG ambience. INNOVATION AND CONCLUSION Our results suggest for the first time that probucol ameliorates renal damage in DN by epigenetically suppressing p66Shc expression via the AMPK-SIRT1-AcH3 pathway.
Collapse
Affiliation(s)
- Shikun Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China; Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Chao Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Ming Zhan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Xiaofen Xiong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Xuejing Zhu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Chun Hu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Zhiguang Zhou
- Diabetes Center, and Institute of Metabolism and Endocrinology, Key Laboratory of Diabetes Immunology, Ministry of Education, China
| | - Yashpal S Kanwar
- Department of Pathology & Medicine, Northwestern University, Chicago, USA
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China.
| |
Collapse
|
34
|
Xiong XF, Yang Y, Chen X, Zhu X, Hu C, Han Y, Zhao L, Liu F, Sun L. Red cell distribution width as a significant indicator of medication and prognosis in type 2 diabetic patients. Sci Rep 2017; 7:2709. [PMID: 28578411 PMCID: PMC5457426 DOI: 10.1038/s41598-017-02904-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/20/2017] [Indexed: 01/18/2023] Open
Abstract
Whether red cell distribution width (RDW) can be a potential indicator for diabetic nephropathy (DN) is unknown. A total of 809 type 2 diabetes mellitus (T2D) patients were divided into 4 groups according to the quartiles (Q) of the RDW (%): Q1 ≤ 12.4 (n = 229), 12.4 < Q2 ≤ 12.9 (n = 202), 12.9 < Q3 < 13.5 (n = 168), Q4 ≥ 13.5 (n = 210). Results showed that the levels in Q4 group was higher in age, disease duration, systolic blood pressure, blood urea nitrogen, creatinine, uric acid and proteinuria but lower in hemoglobin, serum albumin and glycosylated hemoglobin compared to Q1 group. Furthermore, the incidences of DN, diabetic peripheral neuropathy, hypertension and coronary heart disease in the Q3 or Q4 group were higher compared to Q1 group. Medications including calcium channel blockers and antiplatelet therapy also showed higher frequencies in Q3 or Q4 group compared to Q1. Logistic regression indicated that the antiplatelet therapy (OR = 2.065), hypertension (OR = 2.819), creatinine (OR = 4.473) and proteinuria (OR = 2.085) were positively associated with level of Q4 group, but higher hemoglobin (OR = 0.021) and serum Ca2+ (OR = 0.178) were negatively associated with Q4. This data suggest that high level of RDW in T2D patients indicates a higher risk and a poor prognosis for DN.
Collapse
Affiliation(s)
- Xiao-Fen Xiong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yuan Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xianghui Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xuejing Zhu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chun Hu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Li Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|