1
|
Mazzieri A, Porcellati F, Timio F, Reboldi G. Molecular Targets of Novel Therapeutics for Diabetic Kidney Disease: A New Era of Nephroprotection. Int J Mol Sci 2024; 25:3969. [PMID: 38612779 PMCID: PMC11012439 DOI: 10.3390/ijms25073969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Diabetic kidney disease (DKD) is a chronic microvascular complication in patients with diabetes mellitus (DM) and the leading cause of end-stage kidney disease (ESKD). Although glomerulosclerosis, tubular injury and interstitial fibrosis are typical damages of DKD, the interplay of different processes (metabolic factors, oxidative stress, inflammatory pathway, fibrotic signaling, and hemodynamic mechanisms) appears to drive the onset and progression of DKD. A growing understanding of the pathogenetic mechanisms, and the development of new therapeutics, is opening the way for a new era of nephroprotection based on precision-medicine approaches. This review summarizes the therapeutic options linked to specific molecular mechanisms of DKD, including renin-angiotensin-aldosterone system blockers, SGLT2 inhibitors, mineralocorticoid receptor antagonists, glucagon-like peptide-1 receptor agonists, endothelin receptor antagonists, and aldosterone synthase inhibitors. In a new era of nephroprotection, these drugs, as pillars of personalized medicine, can improve renal outcomes and enhance the quality of life for individuals with DKD.
Collapse
Affiliation(s)
- Alessio Mazzieri
- Diabetes Clinic, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (A.M.), (F.P.)
| | - Francesca Porcellati
- Diabetes Clinic, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (A.M.), (F.P.)
| | - Francesca Timio
- Division of Nephrology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| | - Gianpaolo Reboldi
- Division of Nephrology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy;
| |
Collapse
|
2
|
Zhu Q, Li G, Ma L, Chen B, Zhang D, Gao J, Deng S, Chen Y. Virgin Camellia Seed Oil Improves Glycolipid Metabolism in the Kidney of High Fat-Fed Rats through AMPK-SREBP Pathway. Nutrients 2023; 15:4888. [PMID: 38068746 PMCID: PMC10708295 DOI: 10.3390/nu15234888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Camellia seed oil (CO) is used as edible oil in southern China because of its excellent fatty acid composition and abundant bioactive compounds. Chronic kidney disease (CKD) is one of the most common chronic degenerative diseases in China, and active compounds in vegetable oil, like virgin olive oil, have been demonstrated to be efficacious in the management of CKD. In this study, virgin CO was refined using a standard process. The refining had minimal impact on the fatty acid composition, but significantly reduced the presence of bioactive compounds like polyphenols in CO. Sprague-Dawley (SD) rats fed with high fat diet (Group G) were treated with either virgin (Group Z) or refined CO (Group R). The oral administration of CO alleviated lipid accumulation and decreased body and kidney weight gain. Furthermore, treatment with virgin CO increased the renal ATP content. The renal expression levels of AMPK and key enzymes involved in fatty acid oxidation (CPT-1 and ACOX1) and glycolysis (HK, PFK, PK and GAPDH) were up-regulated in Group Z, thereby enhancing the ATP production. Virgin CO treatment downregulated the expression level of SREBP2 and its downstream target genes, such as ACC, FAS, and HMGCR, which reduced lipid synthesis. These findings indicate that virgin CO improves glycolipid metabolism and restores energy homeostasis in the kidneys of rats fed with a high-fat diet by modulating the AMPK-SREBP-signaling pathway, suggesting the potential of active compounds in virgin CO for managing the renal failure associated with glycolipid dysmetabolism.
Collapse
Affiliation(s)
- Qinhe Zhu
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Guihui Li
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Li Ma
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
| | - Bolin Chen
- Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Dawei Zhang
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Jing Gao
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
| | - Senwen Deng
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yongzhong Chen
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
| |
Collapse
|
3
|
Alshammari GM, Al-Ayed MS, Abdelhalim MA, Al-Harbi LN, Yahya MA. Effects of Antioxidant Combinations on the Renal Toxicity Induced Rats by Gold Nanoparticles. Molecules 2023; 28:molecules28041879. [PMID: 36838869 PMCID: PMC9959587 DOI: 10.3390/molecules28041879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
This study investigated some possible mechanisms underlying the nephrotoxic effect of gold nanoparticles (AuNPs) in rats and compared the protective effects of selected known antioxidants-namely, melanin, quercetin (QUR), and α-lipoic acid (α-LA). Rats were divided into five treatment groups (eight rats per group): control, AuNPs (50 nm), AuNPs + melanin (100 mg/kg), AuNPs + QUR (200 mg/kg), and AuNPs + α-LA (200 mg/kg). All treatments were administered i.p., daily, for 30 days. AuNPs promoted renal glomerular and tubular damage and impaired kidney function, as indicated by the higher serum levels of creatinine (Cr), urinary flow, and urea and albumin/Cr ratio. They also induced oxidative stress by promoting mitochondrial permeability transition pore (mtPTP) opening, the expression of NOX4, increasing levels of malondialdehyde (MDA), and suppressing glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT). In addition, AuNPs induced renal inflammation and apoptosis, as evidenced by the increase in the total mRNA and the cytoplasmic and nuclear levels of NF-κB, mRNA levels of Bax and caspase-3, and levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Treatment with melanin, QUR, and α-lipoic acid (α-LA) prevented the majority of these renal damage effects of AuNPs and improved kidney structure and function, with QUR being the most powerful. In conclusion, in rats, AuNPs impair kidney function by provoking oxidative stress, inflammation, and apoptosis by suppressing antioxidants, promoting mitochondrial uncoupling, activating NF-κB, and upregulating NOX4. However, QUR remains the most powerful drug to alleviate this toxicity by reversing all of these mechanisms.
Collapse
Affiliation(s)
- Ghedeir M. Alshammari
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence:
| | - Mohammed S. Al-Ayed
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Anwar Abdelhalim
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Abdo Yahya
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
4
|
Molecular Mechanistic Pathways Targeted by Natural Compounds in the Prevention and Treatment of Diabetic Kidney Disease. Molecules 2022; 27:molecules27196221. [PMID: 36234757 PMCID: PMC9571643 DOI: 10.3390/molecules27196221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 12/03/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common complications of diabetes, and its prevalence is still growing rapidly. However, the efficient therapies for this kidney disease are still limited. The pathogenesis of DKD involves glucotoxicity, lipotoxicity, inflammation, oxidative stress, and renal fibrosis. Glucotoxicity and lipotoxicity can cause oxidative stress, which can lead to inflammation and aggravate renal fibrosis. In this review, we have focused on in vitro and in vivo experiments to investigate the mechanistic pathways by which natural compounds exert their effects against the progression of DKD. The accumulated and collected data revealed that some natural compounds could regulate inflammation, oxidative stress, renal fibrosis, and activate autophagy, thereby protecting the kidney. The main pathways targeted by these reviewed compounds include the Nrf2 signaling pathway, NF-κB signaling pathway, TGF-β signaling pathway, NLRP3 inflammasome, autophagy, glycolipid metabolism and ER stress. This review presented an updated overview of the potential benefits of these natural compounds for the prevention and treatment of DKD progression, aimed to provide new potential therapeutic lead compounds and references for the innovative drug development and clinical treatment of DKD.
Collapse
|
5
|
Liu T, Li CY, Chen H, Liu J, Zhong LL, Tang MM, Wang WB, Huang JP, Jiang XS. tBHQ attenuates podocyte injury in diabetic nephropathy by inhibiting NADPH oxidase-derived ROS generation via the Nrf2/HO-1 signalling pathway. Heliyon 2022; 8:e10515. [PMID: 36119860 PMCID: PMC9479023 DOI: 10.1016/j.heliyon.2022.e10515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/03/2022] [Accepted: 08/26/2022] [Indexed: 12/03/2022] Open
Abstract
Aims Oxidative stress plays a crucial role in podocyte injury in diabetic nephropathy (DN). tert-Butylhydroquinone (tBHQ) is an activator of Nrf2 that exerts protective effects in diabetic mice, but the underlying mechanism of tBHQ in the podocytes of DN is not fully understood. Materials and methods A high glucose (HG)-induced HK2 cell model and streptozotocin-induced rat model of DN were established and treated with tBHQ or apocynin. The expression levels of Nrf2, HO-1, NOX2 and NOX4 were determined by Western blot or immunohistochemical staining. The level of oxidative stress in podocytes or kidney tissues was assessed using DCFH-DA or dihydroethidium (DHE) staining. Cell injury was assessed by F-actin staining and flow cytometry analysis. Key findings We showed that HG treatment increased the expressions of NOX2 and NOX4 and enhanced ROS production in podocytes. Inhibition of NADPH oxidase activity by apocynin dramatically attenuated HG-induced ROS production and further alleviated cell injury and apoptosis in podocytes. Moreover, we found that HG inhibited the Nrf2/HO-1 signalling pathway in podocytes; however, tBHQ treatment significantly activated the Nrf2 signalling pathway, inhibited NADPH oxidase activity, and attenuated ROS production and cell injury in HG-treated podocytes. Furthermore, we observed that tBHQ treatment partially attenuated renal injury, activated the Nrf2 signalling pathway, inhibited NADPH oxidase activity and reduced ROS generation in the kidneys of STZ-induced diabetic rats. Significance These results suggest that tBHQ exerts a protective role in hyperglycaemia-induced podocyte injury, and that the potential protective mechanism of tBHQ involves inhibiting NADPH oxidase-derived ROS generation by activating the Nrf2/HO-1 signalling pathway.
Collapse
Affiliation(s)
- Ting Liu
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Chang-Yan Li
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Hao Chen
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Juan Liu
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Li-Li Zhong
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Ming-Min Tang
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Wen-Bo Wang
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Jin-Ping Huang
- Department of Nephrology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Xu-Shun Jiang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| |
Collapse
|
6
|
Wang Y, Gao L. Inflammation and Cardiovascular Disease Associated With Hemodialysis for End-Stage Renal Disease. Front Pharmacol 2022; 13:800950. [PMID: 35222026 PMCID: PMC8867697 DOI: 10.3389/fphar.2022.800950] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/10/2022] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) and cardiac insufficiency often co-exist, particularly in uremic patients on hemodialysis (HD). The occurrence of abnormal renal function in patients with cardiac insufficiency is often indicative of a poor prognosis. It has long been established that in patients with cardiac insufficiency, poorer renal function tends to indicate poorer cardiac mechanics, including left atrial reserve strain, left ventricular longitudinal strain, and right ventricular free wall strain (Unger et al., Eur J Heart Fail, 2016, 18(1), 103-12). Similarly, patients with chronic kidney disease, particularly uremic patients on HD, often have cardiovascular complications in addition to abnormal endothelial function with volume overload, persistent inflammatory states, calcium overload, and imbalances in redox responses. Cardiac insufficiency due to uremia is therefore mainly due to multifaceted non-specific pathological changes rather than pure renal insufficiency. Several studies have shown that the risk of adverse cardiovascular events is greatly increased and persistent in all patients treated with HD, especially in those who have just started HD treatment. Inflammation, as an important intersection between CKD and cardiovascular disease, is involved in the development of cardiovascular complications in patients with CKD and is indicative of prognosis (Chan et al., Eur Heart J, 2021, 42(13), 1244-1253). Therefore, only by understanding the mechanisms underlying the sequential development of inflammation in CKD patients and breaking the vicious circle between inflammation-mediated renal and cardiac insufficiency is it possible to improve the prognosis of patients with end-stage renal disease (ESRD). This review highlights the mechanisms of inflammation and the oxidative stress that co-exists with inflammation in uremic patients on dialysis, as well as the mechanisms of cardiovascular complications in the inflammatory state, and provides clinical recommendations for the anti-inflammatory treatment of cardiovascular complications in such patients.
Collapse
Affiliation(s)
| | - Lu Gao
- Department of Cardiovascular Centre, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
7
|
Zhao W, Zhou L, Novák P, Shi X, Lin CB, Zhu X, Yin K. Metabolic Dysfunction in the Regulation of the NLRP3 Inflammasome Activation: A Potential Target for Diabetic Nephropathy. J Diabetes Res 2022; 2022:2193768. [PMID: 35719709 PMCID: PMC9203236 DOI: 10.1155/2022/2193768] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
Metabolic dysfunction plays a key role in the development of diabetic nephropathy (DN). However, the exact effects and mechanisms are still unclear. The pyrin domain-containing protein 3 (NLRP3) inflammasome, a member of the nod-like receptor family, is considered a crucial inflammatory regulator and plays important roles in the progress of DN. A growing body of evidence suggests that high glucose, high fat, or other metabolite disorders can abnormally activate the NLRP3 inflammasome. Thus, in this review, we discuss the potential function of abnormal metabolites such as saturated fatty acids (SFAs), cholesterol crystals, uric acid (UA), and homocysteine in the NLRP3 inflammasome activation and explain the potential function of metabolic dysfunction regulation of NLRP3 activation in the progress of DN via regulation of inflammatory response and renal interstitial fibrosis (RIF). In addition, the potential mechanisms of metabolism-related drugs, such as metformin and sodium glucose cotransporter (SGLT2) inhibitors, which have served as the suppressors of the NLRP3 inflammasomes, in DN, are also discussed. A better understanding of NLRP3 inflammasome activation in abnormal metabolic microenvironment may provide new insights for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Wenli Zhao
- Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Xian Shi
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Chuang Biao Lin
- Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Kai Yin
- Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, China
| |
Collapse
|
8
|
Jiang T, Bao Y, Su H, Zheng R, Cao L. Mechanisms of Chinese Herbal Medicines for Diabetic Nephropathy Fibrosis Treatment. INTEGRATIVE MEDICINE IN NEPHROLOGY AND ANDROLOGY 2022; 9. [PMCID: PMC9549772 DOI: 10.4103/2773-0387.353727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Diabetic nephropathy (DN) is a severe microvascular complication of diabetes mellitus that is one of the main causes of end-stage renal disease, causing considerable health problems as well as significant financial burden worldwide. The pathological features of DN include loss of normal nephrons, massive fibroblast and myofibroblast hyperplasia, accumulation of extracellular matrix proteins, thickening of the basement membrane, and tubulointerstitial fibrosis. Renal fibrosis is a final and critical pathological change in DN. Although progress has been made in understanding the pathogenesis of DN fibrosis, current conventional treatment strategies may not be completely effective in preventing the disease’s progression. Traditionally, Chinese herbal medicines (CHMs) composed of natural ingredients have been used for symptomatic relief of DN. Increasing numbers of studies have confirmed that CHMs can exert a renoprotective effect in DN, and antifibrosis has been identified as a key mechanism. In this review, we summarize the antifibrotic efficacy of CHM preparations, single herbal medicines, and their bioactive compounds based on their effects on diminishing the inflammatory response and oxidative stress, regulating transforming growth factor, preventing epithelial-mesenchymal transition, and modulating microRNAs. We intend to provide patients of DN with therapeutic interventions that are complementary to existing options.
Collapse
Affiliation(s)
- Tong Jiang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Yuhang Bao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Hong Su
- Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Rendong Zheng
- Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, China,Address for correspondence: Prof. Rendong Zheng, Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, China. E-mail:
Prof. Lin Cao, Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, China E-mail:
| | - Lin Cao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu Province, China,Address for correspondence: Prof. Rendong Zheng, Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, China. E-mail:
Prof. Lin Cao, Department of Endocrinology and Metabolism, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu Province, China E-mail:
| |
Collapse
|
9
|
Ebert T, Neytchev O, Witasp A, Kublickiene K, Stenvinkel P, Shiels PG. Inflammation and Oxidative Stress in Chronic Kidney Disease and Dialysis Patients. Antioxid Redox Signal 2021; 35:1426-1448. [PMID: 34006115 DOI: 10.1089/ars.2020.8184] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Chronic kidney disease (CKD) can be regarded as a burden of lifestyle disease that shares common underpinning features and risk factors with the aging process; it is a complex constituted by several adverse components, including chronic inflammation, oxidative stress, early vascular aging, and cellular senescence. Recent Advances: A systemic approach to tackle CKD, based on mitigating the associated inflammatory, cell stress, and damage processes, has the potential to attenuate the effects of CKD, but it also preempts the development and progression of associated morbidities. In effect, this will enhance health span and compress the period of morbidity. Pharmacological, nutritional, and potentially lifestyle-based interventions are promising therapeutic avenues to achieve such a goal. Critical Issues: In the present review, currents concepts of inflammation and oxidative damage as key patho-mechanisms in CKD are addressed. In particular, potential beneficial but also adverse effects of different systemic interventions in patients with CKD are discussed. Future Directions: Senotherapeutics, the nuclear factor erythroid 2-related factor 2-kelch-like ECH-associated protein 1 (NRF2-KEAP1) signaling pathway, the endocrine klotho axis, inhibitors of the sodium-glucose cotransporter 2 (SGLT2), and live bio-therapeutics have the potential to reduce the burden of CKD and improve quality of life, as well as morbidity and mortality, in this fragile high-risk patient group. Antioxid. Redox Signal. 35, 1426-1448.
Collapse
Affiliation(s)
- Thomas Ebert
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ognian Neytchev
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Anna Witasp
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
10
|
Liu D, Yu J, Xie J, Zhang Z, Tang C, Yu T, Chen S, Hong Z, Wang C. PbAc Triggers Oxidation and Apoptosis via the PKA Pathway in NRK-52E Cells. Biol Trace Elem Res 2021; 199:2687-2694. [PMID: 32926327 DOI: 10.1007/s12011-020-02378-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/06/2020] [Indexed: 01/28/2023]
Abstract
This study aimed to investigate the mechanism of the lead exposure-induced oxidative stress and apoptosis of renal tubular epithelial cells. We explored the effects of lead acetate (PbAc) on the oxidation and apoptosis of renal proximal tubular cells (NRK-52E) through in vitro experiments. Results showed that PbAc induced dose-dependent reactive oxygen species (ROS) accumulation in NRK-52E cells, and the activities of superoxide dismutase (SOD) and glutathione (GSH) decreased, whereas the malondialdehyde (MDA) content increased. Under the exposure of 40 and 80 μM PbAc, the mRNA level of B cell lymphoma-2 (Bcl-2) in the cells decreased, the mRNA levels of Bcl-2-associated X protein (Bax) and caspase-3 increased, and apoptosis was obvious. Furthermore, the nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) activity was enhanced by PbAc in a dose-dependent manner. The mRNA levels of protein kinase A (PKA) were upregulated by PbAc. H-89, a PKA inhibitor, suppressed PKA activation, ROS accumulation, and Nox4 activity in NRK-52E cells. Our results indicated that PbAc potentially stimulated oxidative stress and apoptosis in NRK-52E cells by increasing Nox4-dependent ROS production via the PKA signaling pathway.
Collapse
Affiliation(s)
- Duanya Liu
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jun Yu
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Jie Xie
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Zhaoyu Zhang
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Caoli Tang
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Tianmei Yu
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Shouni Chen
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Zhidan Hong
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Chunhong Wang
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
11
|
Wahba NS, Ghareib SA, Abdel-Ghany RH, Abdel-Aal M, Alsemeh AE. Vitamin D3 potentiates the nephroprotective effects of metformin in a rat model of metabolic syndrome: role of AMPK/SIRT1 activation and DPP-4 inhibition. Can J Physiol Pharmacol 2021; 99:685-697. [PMID: 33108744 DOI: 10.1139/cjpp-2020-0435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The current study aimed to investigate the molecular mechanisms of metformin and vitamin D3-induced nephroprotection in a metabolic syndrome (MetS) rat model, evaluating the capacity of vitamin D3 to potentiate metformin action. MetS was induced by 10% fructose in drinking water and 3% salt in the diet. After 6 weeks, serum lipid profile and uric acid were measured, an oral glucose tolerance test (OGTT) was performed, and kidney function was investigated. In conjunction with the same concentrations of fructose and salt feeding, MetS rats with significant weight gain, dyslipidemia, hyperuricemia, and dysglycemia were treated orally with metformin (200 mg/kg), vitamin D3 (10 µg/kg), or both daily for 6 weeks. At the end of the study period, anthropometrical parameters were recorded, OGTT was reperformed, urine and blood samples were collected, and tissue samples were harvested at sacrifice. MetS rats showed dramatically declined renal function, enhanced intrarenal oxidative stress and inflammation, and extravagant renal histopathological damage with interstitial fibrosis. Metformin and vitamin D3 significantly reversed all the aforementioned deleterious effects in MetS rats. The study has verified the nephroprotective effects of metformin and vitamin D3 in MetS, accentuating the critical role of AMP-activated protein kinase/sirtuin-1 activation and dipeptidyl peptidase-4 inhibition. Given the synergistic effects of the combination, vitamin D3 is worth being investigated as an additional therapeutic agent for preventing MetS-induced nephropathy.
Collapse
Affiliation(s)
- Nehal S Wahba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Salah A Ghareib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Rasha H Abdel-Ghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed Abdel-Aal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Amira E Alsemeh
- Department of Anatomy and Embryology, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
12
|
Lee H, Jose PA. Coordinated Contribution of NADPH Oxidase- and Mitochondria-Derived Reactive Oxygen Species in Metabolic Syndrome and Its Implication in Renal Dysfunction. Front Pharmacol 2021; 12:670076. [PMID: 34017260 PMCID: PMC8129499 DOI: 10.3389/fphar.2021.670076] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Metabolic syndrome (MetS), a complex of interrelated risk factors for cardiovascular disease and diabetes, is comprised of central obesity (increased waist circumference), hyperglycemia, dyslipidemia (high triglyceride blood levels, low high-density lipoprotein blood levels), and increased blood pressure. Oxidative stress, caused by the imbalance between pro-oxidant and endogenous antioxidant systems, is the primary pathological basis of MetS. The major sources of reactive oxygen species (ROS) associated with MetS are nicotinamide-adenine dinucleotide phosphate (NADPH) oxidases and mitochondria. In this review, we summarize the current knowledge regarding the generation of ROS from NADPH oxidases and mitochondria, discuss the NADPH oxidase- and mitochondria-derived ROS signaling and pathophysiological effects, and the interplay between these two major sources of ROS, which leads to chronic inflammation, adipocyte proliferation, insulin resistance, and other metabolic abnormalities. The mechanisms linking MetS and chronic kidney disease are not well known. The role of NADPH oxidases and mitochondria in renal injury in the setting of MetS, particularly the influence of the pyruvate dehydrogenase complex in oxidative stress, inflammation, and subsequent renal injury, is highlighted. Understanding the molecular mechanism(s) underlying MetS may lead to novel therapeutic approaches by targeting the pyruvate dehydrogenase complex in MetS and prevent its sequelae of chronic cardiovascular and renal diseases.
Collapse
Affiliation(s)
- Hewang Lee
- Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Pedro A Jose
- Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States.,Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
13
|
Yang YY, Deng RR, Chen Z, Yao LY, Yang XD, Xiang DX. Piperazine ferulate attenuates high glucose‑induced mesangial cell injury via the regulation of p66 Shc. Mol Med Rep 2021; 23:374. [PMID: 33760157 PMCID: PMC7985999 DOI: 10.3892/mmr.2021.12013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Diabetic nephropathy (DN) is a severe microvascular complication of diabetes. Hyperglycemia-induced glomerular mesangial cells injury is associated with microvascular damage, which is an important step in the development of DN. Piperazine ferulate (PF) has been reported to exert protective effects against the progression of DN. However, whether PF prevents high glucose (HG)-induced mesangial cell injury remains unknown. The aim of the present study was to investigate the effects of PF on HG-induced mesangial cell injury and to elucidate the underlying mechanisms. Protein and mRNA expression levels were determined via western blot analysis and reverse transcription-quantitative PCR, respectively. IL-6 and TNF-α levels were measured using ELISA. Reactive oxygen species levels and NF-κB p65 nuclear translation were determined via immunofluorescence analysis. Apoptosis was assessed by measuring lactate dehydrogenase (LDH) release, as well as using MTT and flow cytometric assays. The mitochondrial membrane potential of mesangial cells was determined using the JC-1 kit. The results revealed that LDH release were increased; however, cell viability and mitochondrial membrane potential were decreased in the HG group compared with the control group. These changes were inhibited after the mesangial cells were treated with PF. Moreover, PF significantly inhibited the HG-induced production of inflammatory cytokines and the activation of NF-κB in mesangial cells. PF also attenuated the HG-induced upregulation of the expression levels of fibronectin and collagen 4A1. Furthermore, the overexpression of p66Src homology/collagen (Shc) abolished the protective effect of PF on HG-induced mesangial cell injury. In vivo experiments revealed that PF inhibited the activation of inflammatory signaling pathways, glomerular cell apoptosis and mesangial matrix expansion in diabetic mice. Collectively, the present findings demonstrated that PF attenuated HG-induced mesangial cells injury by inhibiting p66Shc.
Collapse
Affiliation(s)
- Yong-Yu Yang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Rong-Rong Deng
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhuo Chen
- Department of Geriatrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Liang-Yuan Yao
- Hunan Provincial Engineering Research Central of Translational Medical and Innovative Drug, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Xi-Ding Yang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Da-Xiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
14
|
Sugahara M, Pak WLW, Tanaka T, Tang SCW, Nangaku M. Update on diagnosis, pathophysiology, and management of diabetic kidney disease. Nephrology (Carlton) 2021; 26:491-500. [PMID: 33550672 DOI: 10.1111/nep.13860] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022]
Abstract
Diabetic kidney disease (DKD) is a chronic complication of diabetes mellitus which may eventually lead to end-stage kidney disease (ESKD). Despite improvements in glycaemic control and blood pressure management with renin-angiotensin-aldosterone system (RAAS) blockade, the current therapy cannot completely halt DKD progression to ESKD in some patients. DKD is a heterogeneous disease entity in terms of its clinical manifestations, histopathology and the rate of progression, which makes it difficult to develop effective therapeutics. It was formerly considered that albuminuria preceded kidney function decline in DKD, but recent epidemiological studies revealed that a distinct group of patients presented kidney dysfunction without developing albuminuria. Other comorbidities, such as hypertension, obesity and gout, also affect the clinical course of DKD. The pathophysiology of DKD is complex and multifactorial, involving both metabolic and haemodynamic factors. These induce activation of intracellular signalling pathways, oxidative stress, hypoxia, dysregulated autophagy and epigenetic changes, which result in kidney inflammation and fibrosis. Recently, two groups of antidiabetic drugs, sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists, were demonstrated to provide renoprotection on top of their glucose-lowering effects. Several other therapeutic agents are also being developed and evaluated in clinical trials.
Collapse
Affiliation(s)
- Mai Sugahara
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Japan
| | - Wai Lun Will Pak
- Renal Unit, Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Japan
| | - Sydney C W Tang
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Japan
| |
Collapse
|
15
|
Srivastava SP, Kanasaki K, Goodwin JE. Loss of Mitochondrial Control Impacts Renal Health. Front Pharmacol 2020; 11:543973. [PMID: 33362536 PMCID: PMC7756079 DOI: 10.3389/fphar.2020.543973] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Disruption of mitochondrial biosynthesis or dynamics, or loss of control over mitochondrial regulation leads to a significant alteration in fuel preference and metabolic shifts that potentially affect the health of kidney cells. Mitochondria regulate metabolic networks which affect multiple cellular processes. Indeed, mitochondria have established themselves as therapeutic targets in several diseases. The importance of mitochondria in regulating the pathogenesis of several diseases has been recognized, however, there is limited understanding of mitochondrial biology in the kidney. This review provides an overview of mitochondrial dysfunction in kidney diseases. We describe the importance of mitochondria and mitochondrial sirtuins in the regulation of renal metabolic shifts in diverse cells types, and review this loss of control leads to increased cell-to-cell transdifferentiation processes and myofibroblast-metabolic shifts, which affect the pathophysiology of several kidney diseases. In addition, we examine mitochondrial-targeted therapeutic agents that offer potential leads in combating kidney diseases.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Keizo Kanasaki
- Internal Medicine 1, Shimane University Faculty of Medicine, Izumo, Japan
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
16
|
Inhibition of RNA-binding protein HuR reduces glomerulosclerosis in experimental nephritis. Clin Sci (Lond) 2020; 134:1433-1448. [PMID: 32478392 PMCID: PMC8086301 DOI: 10.1042/cs20200193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022]
Abstract
Recent identification of an RNA-binding protein (HuR) that regulates mRNA turnover and translation of numerous transcripts via binding to an ARE in their 3′-UTR involved in inflammation and is abnormally elevated in varied kidney diseases offers a novel target for the treatment of renal inflammation and subsequent fibrosis. Thus, we hypothesized that treatment with a selective inhibition of HuR function with a small molecule, KH-3, would down-regulate HuR-targeted proinflammatory transcripts thereby improving glomerulosclerosis in experimental nephritis, where glomerular cellular HuR is elevated. Three experimental groups included normal and diseased rats treated with or without KH-3. Disease was induced by the monoclonal anti-Thy 1.1 antibody. KH-3 was given via daily intraperitoneal injection from day 1 after disease induction to day 5 at the dose of 50 mg/kg BW/day. At day 6, diseased animals treated with KH-3 showed significant reduction in glomerular HuR levels, proteinuria, podocyte injury determined by ameliorated podocyte loss and podocin expression, glomerular staining for periodic acid-Schiff positive extracellular matrix proteins, fibronectin and collagen IV and mRNA and protein levels of profibrotic markers, compared with untreated disease rats. KH-3 treatment also reduced disease-induced increases in renal TGFβ1 and PAI-1 transcripts. Additionally, a marked increase in renal NF-κB-p65, Nox4, and glomerular macrophage cell infiltration observed in disease control group was largely reversed by KH-3 treatment. These results strongly support our hypothesis that down-regulation of HuR function with KH-3 has therapeutic potential for reversing glomerulosclerosis by reducing abundance of pro-inflammatory transcripts and related inflammation.
Collapse
|
17
|
Jayachandran I, Sundararajan S, Venkatesan S, Paadukaana S, Balasubramanyam M, Mohan V, Manickam N. Asymmetric dimethylarginine (ADMA) accelerates renal cell fibrosis under high glucose condition through NOX4/ROS/ERK signaling pathway. Sci Rep 2020; 10:16005. [PMID: 32994511 PMCID: PMC7525240 DOI: 10.1038/s41598-020-72943-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
We previously reported that the circulatory level of Asymmetric dimethylarginine (ADMA), an endogenous competitive inhibitor of nitric oxide synthase, was increased in diabetic kidney disease patients. However, the mechanism and the role of ADMA in diabetic kidney injury remain unclear. Hence, our principal aim is to investigate the causal role of ADMA in the progression of renal cell fibrosis under high glucose (HG) treatment and to delineate its signaling alterations in kidney cell injury. High Glucose/ADMA significantly increased fibrotic events including cell migration, invasion and proliferation along with fibrotic markers in the renal cells; whereas ADMA inhibition reversed the renal cell fibrosis. To delineate the central role of ADMA induced fibrotic signaling pathway and its downstream signaling, we analysed the expression levels of fibrotic markers, NOX4, ROS and ERK activity by using specific inhibitors and genetic manipulation techniques. ADMA stimulated the ROS generation along with a significant increase in NOX4 and ERK activity. Further, we observed that ADMA activated NOX-4 and ERK are involved in the extracellular matrix proteins accumulation. Also, we observed that ADMA induced ERK1/2 phosphorylation was decreased after NOX4 silencing. Our study mechanistically demonstrates that ADMA is involved in the progression of kidney cell injury under high glucose condition by targeting coordinated complex mechanisms involving the NOX4- ROS-ERK pathway.
Collapse
Affiliation(s)
- Isaivani Jayachandran
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India
| | - Saravanakumar Sundararajan
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India
| | - Saravanakumar Venkatesan
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India
| | - Sairaj Paadukaana
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India
| | - Muthuswamy Balasubramanyam
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India
| | - Viswanathan Mohan
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India
| | - Nagaraj Manickam
- Department of Vascular Biology, Madras Diabetes Research Foundation & Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control & ICMR Center for Advanced Research On Diabetes, Chennai, India.
| |
Collapse
|
18
|
Podkowińska A, Formanowicz D. Chronic Kidney Disease as Oxidative Stress- and Inflammatory-Mediated Cardiovascular Disease. Antioxidants (Basel) 2020; 9:E752. [PMID: 32823917 PMCID: PMC7463588 DOI: 10.3390/antiox9080752] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Generating reactive oxygen species (ROS) is necessary for both physiology and pathology. An imbalance between endogenous oxidants and antioxidants causes oxidative stress, contributing to vascular dysfunction. The ROS-induced activation of transcription factors and proinflammatory genes increases inflammation. This phenomenon is of crucial importance in patients with chronic kidney disease (CKD), because atherosclerosis is one of the critical factors of their cardiovascular disease (CVD) and increased mortality. The effect of ROS disrupts the excretory function of each section of the nephron. It prevents the maintenance of intra-systemic homeostasis and leads to the accumulation of metabolic products. Renal regulatory mechanisms, such as tubular glomerular feedback, myogenic reflex in the supplying arteriole, and the renin-angiotensin-aldosterone system, are also affected. It makes it impossible for the kidney to compensate for water-electrolyte and acid-base disturbances, which progress further in the mechanism of positive feedback, leading to a further intensification of oxidative stress. As a result, the progression of CKD is observed, with a spectrum of complications such as malnutrition, calcium phosphate abnormalities, atherosclerosis, and anemia. This review aimed to show the role of oxidative stress and inflammation in renal impairment, with a particular emphasis on its influence on the most common disturbances that accompany CKD.
Collapse
Affiliation(s)
| | - Dorota Formanowicz
- Department of Clinical Biochemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland
| |
Collapse
|
19
|
Yang X, Zhao K, Deng W, Zhao L, Jin H, Mei F, Zhou Y, Li M, Wang W. Apocynin Attenuates Acute Kidney Injury and Inflammation in Rats with Acute Hypertriglyceridemic Pancreatitis. Dig Dis Sci 2020; 65:1735-1747. [PMID: 31617131 DOI: 10.1007/s10620-019-05892-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/05/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Acute hypertriglyceridemic pancreatitis (HTGP) is more likely to be severe and complicated with extrapancreatic organ injury. NOX may be involved in the occurrence and development of high fat acute pancreatitis, but the specific mechanism is not clear. AIMS To investigate the protective effects of apocynin, an inhibitor of NOX, on kidney injury associated with the HTGP and its potential mechanisms in a rat model. METHODS In this study, HTGP rat model was induced by intraperitoneal injection of P-407 and L-Arg in combination. Apocynin was given by subcutaneously injection 30 min before the model was induced. The pancreatic and renal histopathology changes were analyzed. Serum AMY, BUN, Cr levels were measured by the Automatic Biochemistry Analyzer. The expression levels of protein associated with NOX/Akt pathway in the kidney were detected. ROS level in kidney and serum was measured by DHE staining and MDA, SOD kits, respectively. Serum TNF-α and IL-6 were detected by ELISA kits. RESULTS In HTGP group, the levels of serum AMY, BUN, Cr, TNF- α, and IL-6 were significantly increased, and the injury of pancreas and kidney was aggravated. The levels of NOX4, NOX2, ROS, p-Akt, GSK-3β, NF-κB, and TNF-α in the kidney were detected, suggesting that NOX may regulate the activity of downstream p-Akt and GSK-3β by regulating ROS levels, thereby affecting the release of inflammatory mediators and regulating HTGP-related kidney injury. After application of apocynin, the expression of NOX4 and NOX2 and the level of ROS in the kidney were reduced, the release of inflammatory mediators decreased, and the histopathology injury of pancreas and kidney was improved obviously. CONCLUSION NOX may play an important role in HTGP-associated kidney injury through Akt/GSK-3β pathway. Apocynin can significantly downregulate the level of NOX and play a protective role in HTGP-related kidney injury through Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Xiaojia Yang
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China
| | - Kailiang Zhao
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China
| | - Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China
| | - Liang Zhao
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China
| | - Hongzhong Jin
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Fangchao Mei
- Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yu Zhou
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Man Li
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, China.
| |
Collapse
|
20
|
Shen X, Dong X, Han Y, Li Y, Ding S, Zhang H, Sun Z, Yin Y, Li W, Li W. Ginsenoside Rg1 ameliorates glomerular fibrosis during kidney aging by inhibiting NOX4 and NLRP3 inflammasome activation in SAMP8 mice. Int Immunopharmacol 2020; 82:106339. [PMID: 32114413 DOI: 10.1016/j.intimp.2020.106339] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 01/08/2023]
Abstract
Aging is closely related to the progress of renal fibrosis, which eventually results in renal dysfunction. Ginsenoside Rg1 (Rg1) has been reported to have an extensive anti-aging effect. However, the role and mechanism of Rg1 in aging-related renal fibrosis remain unclear. The present study aimed to evaluate the protective effect and mechanism of Rg1 in renal fibrosis during kidney aging in a model of SAMP8 mice. Taking SAMR1 mice as the control group, SAMP8 mice were administered Apocynin (50 mg/kg), Tempol (50 mg/kg), or Rg1 (5, 10 mg/kg) intragastrically for 9 weeks as treatment groups. The results showed that the elevated levels of blood urea nitrogen, serum creatinine and senescence-associated β-galactosidase (β-Gal) were markedly decreased, the glomerular mesangial proliferation was significantly alleviated and the increased levels of collagen IV and TGF-β1 were significantly downregulated by Rg1 in SAMP8 mice. In addition, the generation of ROS and the expression of NADHP oxidase 4 (NOX4) in the renal cortex were significantly reduced by Rg1 treatment. The expression levels of NLRP3 inflammasome-related proteins and the inflammation-related cytokine IL-1β were also inhibited by Rg1 treatment in the SAMP8 mice. These results suggested that Rg1 could delay kidney aging and inhibit aging-related glomerular fibrosis by reducing NOX4-derived ROS generation and downregulating NLRP3 inflammasome expression.
Collapse
Affiliation(s)
- Xiaoyan Shen
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Xianan Dong
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yuli Han
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yan Li
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Shixin Ding
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Han Zhang
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Zhenghao Sun
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yanyan Yin
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Weiping Li
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China; Anqing Medical and Pharmaceutical College, Anqing 246052, Anhui, China.
| | - Weizu Li
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
21
|
Targeting Redox Imbalance as an Approach for Diabetic Kidney Disease. Biomedicines 2020; 8:biomedicines8020040. [PMID: 32098346 PMCID: PMC7167917 DOI: 10.3390/biomedicines8020040] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetic kidney disease (DKD) is a worldwide public health problem. It is the leading cause of end-stage renal disease and is associated with increased mortality from cardiovascular complications. The tight interactions between redox imbalance and the development of DKD are becoming increasingly evident. Numerous cascades, including the polyol and hexosamine pathways have been implicated in the oxidative stress of diabetes patients. However, the precise molecular mechanism by which oxidative stress affects the progression of DKD remains to be elucidated. Given the limited therapeutic options for DKD, it is essential to understand how oxidants and antioxidants are controlled in diabetes and how oxidative stress impacts the progression of renal damage. This review aims to provide an overview of the current status of knowledge regarding the pathological roles of oxidative stress in DKD. Finally, we summarize recent therapeutic approaches to preventing DKD with a focus on the anti-oxidative effects of newly developed anti-hyperglycemic agents.
Collapse
|
22
|
Glezer MG. Antihypertensive Effectiveness of Perindopril Arginine and Indapamide Single-Pill Combination According to Body Mass Index: Findings from the FORSAGE Study. Cardiol Ther 2020; 9:139-152. [PMID: 32008207 PMCID: PMC7237592 DOI: 10.1007/s40119-020-00162-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Indexed: 12/01/2022] Open
Abstract
Introduction Overweight and obesity are increasing worldwide and are associated with an increased risk for cardiovascular disease (CVD). The aim of this study was to examine the burden of CVD risk factors among normal weight, overweight, and obese subjects with hypertension, and to evaluate the effectiveness of switching to a single-pill combination (SPC) of perindopril arginine/indapamide for blood pressure (BP) control in overweight and obese subjects treated in routine clinical practice. Methods FORSAGE was a 3-month, multicenter, observational, open-label study conducted in Russian patients with uncontrolled arterial hypertension under previous antihypertensive therapy. Subjects were switched to the full-dose perindopril arginine 10 mg/indapamide 2.5 mg SPC. BP was assessed at 2 weeks, 1 month, and 3 months, and serum creatinine and general health status at 3 months. The present post hoc analysis of the FORSAGE study results explored the effectiveness of perindopril arginine/indapamide SPC in patients with arterial hypertension with regard to baseline body mass index (BMI): normal (< 25 kg/m2), overweight (25 ≤ BMI < 30 kg/m2), and obese (≥ 30 kg/m2). Results A total of 1969 patients were recruited, but BMI data were available for 1963 patients, two-thirds of whom were women. The distribution of BMI groups was as follows: < 25 kg/m2 (16.7%), overweight (48.7%), and obese (34.7%). Overweight or obese patients had more concomitant diseases such as diabetes mellitus or history of stroke, higher BP levels, serum cholesterol and creatinine, and lower glomerular filtration rates. Switching to perindopril arginine/indapamide SPC was associated with a statistically significant reduction in BP as early as the second week of treatment. At 3 months, systolic blood pressure (SBP)/diastolic blood pressure (DBP) had decreased significantly by 39.3/18.8 mmHg in the normal BMI group, 39.8/18.8 mmHg in overweight, and 39.4/18.7 mmHg in obese groups. The magnitude of the BP reduction was independent of BMI. Achievement of target BP (< 140/90 mmHg) was good in all groups, but lower in obese subjects (70.9%) than in overweight subjects (78.1%) or those with a normal BMI (81.8%) (P < 0.0001 for both comparisons). Conclusions In subjects with uncontrolled BP on existing antihypertensive therapy, switching to perindopril arginine 10 mg/indapamide 2.5 mg was associated with statistically significant decreases in BP and higher rates of target BP achievement in all BMI groups, including more than 70% of overweight and obese patients. Trial Registration ISRCTN ID, ISRCTN14315146 (retrospectively registered 18/11/2019). Electronic supplementary material The online version of this article (10.1007/s40119-020-00162-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M G Glezer
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russia.
| | | |
Collapse
|
23
|
Miguel V, Lamas S. Redox distress in organ fibrosis: The role of noncoding RNAs. OXIDATIVE STRESS 2020:779-820. [DOI: 10.1016/b978-0-12-818606-0.00037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Wahba NS, Abdel-Ghany RH, Ghareib SA, Abdel-Aal M, Alsemeh AE. Vitamin D3 potentiates the renoprotective effects of vildagliptin in a rat model of fructose/salt-induced insulin resistance. Eur J Pharm Sci 2019; 144:105196. [PMID: 31866564 DOI: 10.1016/j.ejps.2019.105196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/18/2019] [Accepted: 12/14/2019] [Indexed: 01/22/2023]
Abstract
Insulin resistance (IR) seemingly plays a role in chronic kidney disease (CKD). The present study has elucidated the crucial interplay of oxidative stress, inflammatory, apoptotic and profibrotic signaling pathways, linking IR to CKD. The study aimed at investigating the pleiotropic nephroprotective effects of either vildagliptin or vitamin D3 in a fructose/salt-induced IR rat model, highlighting the potential molecular mechanisms underlying their action. Another interesting target was to evaluate the potential capacity of vitamin D3 to potentiate the nephroprotective effects of vildagliptin. Indeed, a state of impaired fasting glucose, IR and compensatory hyperinsulinemia, constellating with significant weight gain, atherogenic dyslipidemia and hyperuricemia was established 6 weeks after fructose/salt consumption. IR rats were then treated orally with vildagliptin (10 mg/kg/day), vitamin D3 (10 µg/kg/day) or their combination for a further 6 weeks. By the end of the 12th week, untreated IR rats displayed significantly declined renal function with parallel interwined renal oxidative stress, inflammatory, apoptotic and profibrotic changes, renal histopathological damages and markedly enhanced collagen fiber deposition. Vildagliptin and vitamin D3 reversed hyperuricemia and exerted a plethora of renal anti-oxidant, anti-inflammatory, anti-apoptotic and anti-fibrotic effects. Our study has introduced a new insight into the role of dipeptidyl peptidase-4 inhibition and silent information regulator 1/5'adenosine monophosphate-activated protein kinase activation in the nephroprotective effects of either agent, elucidating their possible crosstalk with renin angiotensin aldosterone system downregulation. Considering the superadditive renoprotective effects evoked by the combination, vitamin D3 is worth being further investigated as an additional therapeutic agent for preventing IR-induced nephropathy.
Collapse
Affiliation(s)
- Nehal S Wahba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Rasha H Abdel-Ghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Salah A Ghareib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed Abdel-Aal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Amira E Alsemeh
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
25
|
Oxidative Stress and Renal Fibrosis: Mechanisms and Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:585-604. [PMID: 31399986 DOI: 10.1007/978-981-13-8871-2_29] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oxidative stress results from the disruption of the redox system marked by a notable overproduction of reactive oxygen species. There are four major sources of reactive oxygen species, including NADPH oxidases, mitochondria, nitric oxide synthases, and xanthine oxidases. It is well known that renal abnormalities trigger the production of reactive oxygen species by diverse mechanisms under various pathologic stimuli, such as acute kidney injury, chronic kidney disease, nephrotic syndrome, and metabolic disturbances. Mutually, accumulating evidences have identified that oxidative stress plays an essential role in tubulointerstitial fibrosis by myofibroblast activation as well as in glomerulosclerosis by mesangial sclerosis, podocyte abnormality, and parietal epithelial cell injury. Given the involvement of oxidative stress in renal fibrosis, therapies targeting oxidative stress seem promising in renal fibrosis management. In this review, we sketch the updated knowledge of the mechanisms of oxidative stress generation during renal diseases, the pathogenic processes of oxidative stress elicited renal fibrosis and treatments targeting oxidative stress during tubulointerstitial fibrosis and glomerulosclerosis.
Collapse
|
26
|
Zhang Q, Conley SM, Li G, Yuan X, Li PL. Rac1 GTPase Inhibition Blocked Podocyte Injury and Glomerular Sclerosis during Hyperhomocysteinemia via Suppression of Nucleotide-Binding Oligomerization Domain-Like Receptor Containing Pyrin Domain 3 Inflammasome Activation. Kidney Blood Press Res 2019; 44:513-532. [PMID: 31266025 PMCID: PMC6800118 DOI: 10.1159/000500457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Elevated homocysteine (Hcy) levels have been shown to activate nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome leading to podocyte dysfunction and glomerular injury. However, it remains unclear how this inflammasome activation in podocytes is a therapeutic target for reversal of glomerular injury and ultimate sclerosis. The present study tested whether inhibition of Rac1 GTPase activity suppresses NLRP3 inflammation activation and thereby blocks podocyte injury induced by elevated Hcy. In cultured podocytes, we found that L-Hcy (the active Hcy form) stimulated the NLRP3 inflammasome formation, as shown by increased colocalization of NLRP3 with apoptosis-associated speck-like protein (ASC) or caspase-1, which was accompanied by increased interleukin-1β production and caspase-1 activity, indicating NLRP3 inflammasome activation. Rac1 activator, uridine triphosphate (UTP), mimicked L-Hcy-induced NLRP3 inflammasome activation, while Rac1 inhibitor NSC23766 blocked it. This Rac1 inhibition also prevented L-Hcy-induced podocyte dysfunction. All these effects were shown to be mediated via lipid raft redox signaling platforms with nicotinamide adenine dinucleotide phosphate oxidase subunits and consequent O2− production. In animal studies, hyperhomocysteinemia (hHcy) induced by folate-free diet was shown to induce NLRP3 inflammasome formation and activation in glomeruli, which was also mimicked by UTP and inhibited by NSC23766 to a comparable level seen in Nlrp3 gene knockout mice. These results together suggest that Rac1 inhibition protects the kidney from hHcy-induced podocyte injury and glomerular sclerosis due to its action to suppress NLRP3 inflammasome activation in podocytes.
Collapse
Affiliation(s)
- Qinghua Zhang
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sabena M Conley
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Guangbi Li
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Xinxu Yuan
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pin-Lan Li
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA,
| |
Collapse
|
27
|
Touyz RM, Anagnostopoulou A, Rios F, Montezano AC, Camargo LL. NOX5: Molecular biology and pathophysiology. Exp Physiol 2019; 104:605-616. [PMID: 30801870 PMCID: PMC6519284 DOI: 10.1113/ep086204] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
NEW FINDINGS What is the topic of this review? This review provides a comprehensive overview of Nox5 from basic biology to human disease and highlights unique features of this Nox isoform What advances does it highlight? Major advances in Nox5 biology relate to crystallization of the molecule and new insights into the pathophysiological role of Nox5. Recent discoveries have unravelled the crystal structure of Nox5, the first Nox isoform to be crystalized. This provides new opportunities to develop drugs or small molecules targeted to Nox5 in an isoform-specific manner, possibly for therapeutic use. Moreover genome wide association studies (GWAS) identified Nox5 as a new blood pressure-associated gene and studies in mice expressing human Nox5 in a cell-specific manner have provided new information about the (patho) physiological role of Nox5 in the cardiovascular system and kidneys. Nox5 seems to be important in the regulation of vascular contraction and kidney function. In cardiovascular disease and diabetic nephropathy, Nox5 activity is increased and this is associated with increased production of reactive oxygen species and oxidative stress implicated in tissue damage. ABSTRACT Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox), comprise seven family members (Nox1-Nox5 and dual oxidase 1 and 2) and are major producers of reactive oxygen species in mammalian cells. Reactive oxygen species are crucially involved in cell signalling and function. All Noxs share structural homology comprising six transmembrane domains with two haem-binding regions and an NADPH-binding region on the intracellular C-terminus, whereas their regulatory systems, mechanisms of activation and tissue distribution differ. This explains the diverse function of Noxs. Of the Noxs, NOX5 is unique in that rodents lack the gene, it is regulated by Ca2+ , it does not require NADPH oxidase subunits for its activation, and it is not glycosylated. NOX5 localizes in the perinuclear and endoplasmic reticulum regions of cells and traffics to the cell membrane upon activation. It is tightly regulated through numerous post-translational modifications and is activated by vasoactive agents, growth factors and pro-inflammatory cytokines. The exact pathophysiological significance of NOX5 remains unclear, but it seems to be important in the physiological regulation of sperm motility, vascular contraction and lymphocyte differentiation, and NOX5 hyperactivation has been implicated in cardiovascular disease, kidney injury and cancer. The field of NOX5 biology is still in its infancy, but with new insights into its biochemistry and cellular regulation, discovery of the NOX5 crystal structure and genome-wide association studies implicating NOX5 in disease, the time is now ripe to advance NOX5 research. This review provides a comprehensive overview of our current understanding of NOX5, from basic biology to human disease, and highlights the unique characteristics of this enigmatic Nox isoform.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Aikaterini Anagnostopoulou
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Francisco Rios
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| | - Livia L. Camargo
- Institute of Cardiovascular and Medical SciencesBHF Glasgow Cardiovascular CentreUniversity of GlasgowGlasgowUK
| |
Collapse
|
28
|
Ostrakhovitch EA, Tabibzadeh S. Homocysteine and age-associated disorders. Ageing Res Rev 2019; 49:144-164. [PMID: 30391754 DOI: 10.1016/j.arr.2018.10.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/30/2018] [Accepted: 10/25/2018] [Indexed: 12/26/2022]
Abstract
There are numerous theories of aging, a process which still seems inevitable. Aging leads to cancer and multi-systemic disorders as well as chronic diseases. Decline in age- associated cellular functions leads to neurodegeneration and cognitive decline that affect the quality of life. Accumulation of damage, mutations, metabolic changes, failure in cellular energy production and clearance of altered proteins over the lifetime, and hyperhomocysteinemia, ultimately result in tissue degeneration. The decline in renal functions, nutritional deficiencies, deregulation of methionine cycle and deficiencies of homocysteine remethylation and transsulfuration cofactors cause elevation of homocysteine with advancing age. Abnormal accumulation of homocysteine is a risk factor of cardiovascular, neurodegenerative and chronic kidney disease. Moreover, approximately 50% of people, aged 65 years and older develop hypertension and are at a high risk of developing cardiovascular insufficiency and incurable neurodegenerative disorders. Increasing evidence suggests inverse relation between cognitive impairment, cerebrovascular and cardiovascular events and renal function. Oxidative stress, inactivation of nitric oxide synthase pathway and mitochondria dysfunction associated with impaired homocysteine metabolism lead to aging tissue degeneration. In this review, we examine impact of high homocysteine levels on changes observed with aging that contribute to development and progression of age associated diseases.
Collapse
Affiliation(s)
- E A Ostrakhovitch
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA, USA.
| | - S Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA, USA.
| |
Collapse
|
29
|
Lazarova D, Shibata S, Ishii I, Zlateva G, Zhelev Z, Aoki I, Bakalova R. Imaging of redox-imbalance and oxidative stress in kidney in vivo, induced by dietary cholesterol. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1573153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Dessislava Lazarova
- Department of Physics, Biophysics and Roentgenology, Medical Faculty, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Sayaka Shibata
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
- Group of Quantum-State Controlled MRI, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
| | - Itsuko Ishii
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Chiba University Hospital, Chiba University, Chiba, Japan
| | - Genoveva Zlateva
- Department of Physics, Biophysics and Roentgenology, Medical Faculty, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Zhivko Zhelev
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
- Department of Electroinduced and Adhesive Properties, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ichio Aoki
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
- Group of Quantum-State Controlled MRI, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
| | - Rumiana Bakalova
- Department of Physics, Biophysics and Roentgenology, Medical Faculty, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
- Group of Quantum-State Controlled MRI, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
| |
Collapse
|
30
|
Jiang Y, Liu J, Zhou Z, Liu K, Liu C. Diosmetin Attenuates Akt Signaling Pathway by Modulating Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells (NF-κB)/Inducible Nitric Oxide Synthase (iNOS) in Streptozotocin (STZ)-Induced Diabetic Nephropathy Mice. Med Sci Monit 2018; 24:7007-7014. [PMID: 30278036 PMCID: PMC6354632 DOI: 10.12659/msm.910764] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background We evaluated the nephroprotective effect of diosmetin in streptozotocin (STZ)-induced diabetic nephropathy (DN) mice. Material/Methods Diabetes was induced by injecting STZ (50 mg/kg) i.p. for 5 days. Biochemical parameters, such as fasting blood glucose, creatinine, BUN in the serum, and albumin in the urine, were determined in STZ-induced DN mice after the 8th week of STZ administration. The level of inflammatory mediators in the serum and oxidative stress parameters in the tissue homogenate was estimated in STZ-induced DN mice. Expressions of Akt, NF-κB, and iNOS in the tissue homogenate were assessed by Western blot analysis. Results Our data reveal that treatment with diosmetin significantly reduces the fasting blood glucose (FBG), serum creatinine, and blood urea nitrogen (BUN) in the serum and albumin in urine compared to the negative control group. Treatment with diosmetin attenuated the altered level of oxidative stress parameters and inflammatory cytokines in the STZ-induced DN mice. Expression of Akt and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) was significantly reduced and inducible nitric oxide synthase (iNOS) was enhanced in the tissue homogenate of diosmetin-treated mice compared to the negative control group. Data from immunohistochemical analysis suggest that the expressions of NF-κB was significantly reduced in tissues of the diosmetin-treated group compared to the negative control group. Conclusions Our study shows that diosmetin protects against renal injury in STZ-induced diabetic nephropathy mice by modulating the Akt/NF-κB/iNOS signaling pathway.
Collapse
Affiliation(s)
- Yingsong Jiang
- Department of Nephrology, Chongqing General Hospital, Chongqing, China (mainland)
| | - Jiguo Liu
- Department of Nephrology, Chongqing General Hospital, Chongqing, China (mainland)
| | - Zemei Zhou
- Department of Nephrology, Chongqing General Hospital, Chongqing, China (mainland)
| | - Ke Liu
- Department of Nephrology, Chongqing General Hospital, Chongqing, China (mainland)
| | - Chun Liu
- Department of Nephrology, Chongqing General Hospital, Chongqing, China (mainland)
| |
Collapse
|
31
|
Thippakorn C, Schaduangrat N, Nantasenamat C. Proteomic and bioinformatic discovery of biomarkers for diabetic nephropathy. EXCLI JOURNAL 2018; 17:312-330. [PMID: 29805343 PMCID: PMC5962897 DOI: 10.17179/excli2018-1150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/12/2018] [Indexed: 11/13/2022]
Abstract
Diabetes is associated with numerous metabolic and vascular risk factors that contribute to a high rate of micro-vascular and macro-vascular disorders leading to mortality and morbidity from diabetic complications. In this case, the major cause of death in overall diabetic patients results from diabetic nephropathy (DN) or renal failure. The risk factors and mechanisms that correspond to the development of DN are not fully understood and so far, no specific and sufficient diagnostic biomarkers are currently available other than micro- or macroalbuminuria. Therefore, this review describes current and novel protein biomarkers in the context of DN as well as probable proteins biomarkers associated with pathological processes for the early stage of DN via proteomics data together with bioinformatics. In addition, the mechanisms involved in early development of diabetic vascular disorders and complications resulting from glucose induced oxidative stress will also be explored.
Collapse
Affiliation(s)
- Chadinee Thippakorn
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
32
|
Alhasson F, Seth RK, Sarkar S, Kimono DA, Albadrani MS, Dattaroy D, Chandrashekaran V, Scott GI, Raychoudhury S, Nagarkatti M, Nagarkatti P, Diehl AM, Chatterjee S. High circulatory leptin mediated NOX-2-peroxynitrite-miR21 axis activate mesangial cells and promotes renal inflammatory pathology in nonalcoholic fatty liver disease. Redox Biol 2018; 17:1-15. [PMID: 29660503 PMCID: PMC6006523 DOI: 10.1016/j.redox.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
High circulatory insulin and leptin followed by underlying inflammation are often ascribed to the ectopic manifestations in non-alcoholic fatty liver disease (NAFLD) but the exact molecular pathways remain unclear. We have shown previously that CYP2E1-mediated oxidative stress and circulating leptin in NAFLD is associated with renal disease severity. Extending the studies, we hypothesized that high circulatory leptin in NAFLD causes renal mesangial cell activation and tubular inflammation via a NOX2 dependent pathway that upregulates proinflammatory miR21. High-fat diet (60% kcal) was used to induce fatty liver phenotype with parallel insulin and leptin resistance. The kidneys were probed for mesangial cell activation and tubular inflammation that showed accelerated NASH phenotype and oxidative stress in the liver. Results showed that NAFLD kidneys had significant increases in α-SMA, a marker of mesangial cell activation, miR21 levels, tyrosine nitration and renal inflammation while they were significantly decreased in leptin and p47 phox knockout mice. Micro RNA21 knockout mice showed decreased tubular immunotoxicity and proinflammatory mediator release. Mechanistically, use of NOX2 siRNA or apocynin,phenyl boronic acid (FBA), DMPO or miR21 antagomir inhibited leptin primed-miR21-mediated mesangial cell activation in vitro suggesting a direct role of leptin-mediated NOX-2 in miR21-mediated mesangial cell activation. Finally, JAK-STAT inhibitor completely abrogated the mesangial cell activation in leptin-primed cells suggesting that leptin signaling in the mesangial cells depended on the JAK-STAT pathway. Taken together the study reports a novel mechanistic pathway of leptin-mediated renal inflammation that is dependent on NOX-2-miR21 axis in ectopic manifestations underlying NAFLD-induced co-morbidities.
Collapse
Affiliation(s)
- Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Sutapa Sarkar
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Diana A Kimono
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Muayad S Albadrani
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Varun Chandrashekaran
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Geoffrey I Scott
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Samir Raychoudhury
- Department of Biology, Chemistry and Environmental Health Science, Benedict College, Columbia, SC 29204, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, NC 27707, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
33
|
Liu M, Zhu Y, Sun Y, Wen Z, Huang S, Ding G, Zhang A, Jia Z, Zhang Y. MnTBAP therapy attenuates the downregulation of sodium transporters in obstructive kidney disease. Oncotarget 2017; 9:394-403. [PMID: 29416622 PMCID: PMC5787475 DOI: 10.18632/oncotarget.23037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/13/2017] [Indexed: 11/25/2022] Open
Abstract
Ureteral obstruction is associated with reduced expressions of renal sodium transporters, which contributes to impaired urinary concentrating capacity. In this study, we employed a synthetic mitochondrial superoxide dismutase 2 (SOD2) mimic MnTBAP to investigate the role of mitochondrial oxidative stress in modulating the sodium transporters in obstructive kidney disease. Following unilateral ureteral obstruction (UUO) for 7 days, a global reduction of sodium transporters including NHE3, NCC, NKCC2, and ENaCα was observed as determined by qRT-PCR, Western Blotting or immunohistochemistry. Among these sodium transporters, the downregulation of NHE3, NCC, and NKCC2 was partially reversed by MnTBAP treatment. In contrast, the reduction of ENaCα was not affected by MnTBAP. The β and γ subunits of ENaC were not significantly altered by ureteral obstruction or MnTBAP therapy. To further confirm the anti-oxidant effect of MnTBAP, we examined the levels of TBARs in the urine collected from the obstructed ureters of UUO mice and bladder of sham mice. As expected, the increment of urinary TBARs in UUO mice was entirely abolished by MnTBAP therapy, indicating an amelioration of oxidative stress. Meantime, we found that three types of SOD were all reduced in obstructed kidneys determined by qRT-PCR, which was unaffected by MnTBAP. Collectively, these results demonstrated an important role of mitochondrial oxidative stress in mediating the downregulation of sodium transporters in obstructive kidney disease.
Collapse
Affiliation(s)
- Mi Liu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yangyang Zhu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Ying Sun
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhaoying Wen
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Guixia Ding
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
34
|
Idelchik MDPS, Begley U, Begley TJ, Melendez JA. Mitochondrial ROS control of cancer. Semin Cancer Biol 2017; 47:57-66. [PMID: 28445781 PMCID: PMC5653465 DOI: 10.1016/j.semcancer.2017.04.005] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 04/07/2017] [Accepted: 04/14/2017] [Indexed: 02/06/2023]
Abstract
Mitochondria serves a primary role in energy maintenance but also function to govern levels of mitochondria-derived reactive oxygen species (mROS). ROS have long been established to play a critical role in tumorigenesis and are now considered to be integral to the regulation of diverse signaling networks that drive proliferation, tumor cell survival and malignant progression. mROS can damage DNA, activate oncogenes, block the function of tumor suppressors and drive migratory signaling. The mitochondrion's oxidant scavenging systems including SOD2, Grx2, GPrx, Trx and TrxR are key of the cellular redox tone. These mitochondrial antioxidant systems serve to tightly control the levels of the primary ROS signaling species, H2O2. The coordinated control of mROS levels is also coupled to the activity of the primary H2O2 consuming enzymes of the mitochondria which are reliant on the epitranscriptomic control of selenocysteine incorporation. This review highlights the interplay between these many oncogenic signaling networks, mROS and the H2O2 emitting and consuming capacity of the mitochondria.
Collapse
Affiliation(s)
- María Del Pilar Sosa Idelchik
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States
| | - Ulrike Begley
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States
| | - Thomas J Begley
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States
| | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, NFE-4313, Albany, NY 12203, United States.
| |
Collapse
|
35
|
Argyropoulos CP, Chen SS, Ng YH, Roumelioti ME, Shaffi K, Singh PP, Tzamaloukas AH. Rediscovering Beta-2 Microglobulin As a Biomarker across the Spectrum of Kidney Diseases. Front Med (Lausanne) 2017; 4:73. [PMID: 28664159 PMCID: PMC5471312 DOI: 10.3389/fmed.2017.00073] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/26/2017] [Indexed: 12/28/2022] Open
Abstract
There is currently an unmet need for better biomarkers across the spectrum of renal diseases. In this paper, we revisit the role of beta-2 microglobulin (β2M) as a biomarker in patients with chronic kidney disease and end-stage renal disease. Prior to reviewing the numerous clinical studies in the area, we describe the basic biology of β2M, focusing in particular on its role in maintaining the serum albumin levels and reclaiming the albumin in tubular fluid through the actions of the neonatal Fc receptor. Disorders of abnormal β2M function arise as a result of altered binding of β2M to its protein cofactors and the clinical manifestations are exemplified by rare human genetic conditions and mice knockouts. We highlight the utility of β2M as a predictor of renal function and clinical outcomes in recent large database studies against predictions made by recently developed whole body population kinetic models. Furthermore, we discuss recent animal data suggesting that contrary to textbook dogma urinary β2M may be a marker for glomerular rather than tubular pathology. We review the existing literature about β2M as a biomarker in patients receiving renal replacement therapy, with particular emphasis on large outcome trials. We note emerging proteomic data suggesting that β2M is a promising marker of chronic allograft nephropathy. Finally, we present data about the role of β2M as a biomarker in a number of non-renal diseases. The goal of this comprehensive review is to direct attention to the multifaceted role of β2M as a biomarker, and its exciting biology in order to propose the next steps required to bring this recently rediscovered biomarker into the twenty-first century.
Collapse
Affiliation(s)
- Christos P Argyropoulos
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Shan Shan Chen
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Yue-Harn Ng
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Maria-Eleni Roumelioti
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Kamran Shaffi
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Pooja P Singh
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Antonios H Tzamaloukas
- Nephrology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States.,Raymond G. Murphy VA Medical Center Albuquerque, Albuquerque, NM, United States
| |
Collapse
|
36
|
Al-Harbi NO, Nadeem A, Ansari MA, Al-Harbi MM, Alotaibi MR, AlSaad AM, Ahmad SF. Psoriasis-like inflammation leads to renal dysfunction via upregulation of NADPH oxidases and inducible nitric oxide synthase. Int Immunopharmacol 2017; 46:1-8. [DOI: 10.1016/j.intimp.2017.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 02/07/2023]
|
37
|
Zheng D, Zhao Y, Liu L, Sun X, Xia Y, Sun L, Xie K. Differential expression profile analysis of PSTK-regulated mRNAs in podocytes. J Cell Biochem 2017; 120:8935-8948. [PMID: 28419530 DOI: 10.1002/jcb.26076] [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: 12/21/2016] [Accepted: 04/14/2018] [Indexed: 11/12/2022]
Abstract
This study aimed to elucidate the precise mechanisms underlying the protective effects of phosphoseryl-tRNA kinase (PSTK) against cisplatin-induced podocyte injury. PSTK overexpression and knockdown vectors were generated and transfected into murine podocyte cells-5. PSTK levels were measured, and transcriptome sequencing was conducted. Differential expression analysis was performed to identify messenger RNAs (mRNAs) that were positively and negatively correlated with PSTK. We selected 10 candidate genes identified via real-time quantitative polymerase chain reaction and Western blot analysis for further analysis. As expected, PSTK levels were significantly higher in PSTK-overexpressing podocytes and significantly lower in PSTK-knockdown podocytes. PSTK overexpression resulted in the upregulation of 122 genes and downregulation of 372 genes in podocytes. On the other hand, PSTK knockdown resulted in the upregulation of 231 genes and downregulation of 445 genes. Furthermore, the analysis revealed that 11 genes were positively correlated with PSTK, whereas 20 genes were negatively correlated with PSTK. The obtained PSTK-regulated genes were primarily involved in molecular function, biological process, and cellular component, as well as the angiogenesis pathway. The Wnt family member 10A levels were significantly higher after PSTK overexpression, but were significantly lower after PSTK knockdown. In addition, Na+/K+ ATPase subunit α-2 and matrix metalloproteinase 9 levels were significantly downregulated after PSTK overexpression, but significantly upregulated upon PSTK knockdown. Cell proliferation was significantly increased upon PSTK overexpression, but significantly decreased upon PSTK suppression. The results of this study not only identified several significant PSTK-regulated genes for further validation, but also provided insights into the mechanisms underlying the protective effects of PSTK on podocytes.
Collapse
Affiliation(s)
- Dong Zheng
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Limin Liu
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xiaodong Sun
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yiyuan Xia
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Lina Sun
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Keming Xie
- Department of Pathology and Pathophysiology, Medical College of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|