1
|
Chen D, Wang S, Sun Z, Zhang X, Chong N, Shu J, Wang Q, Xu Y. Network pharmacology, molecular docking, and experimental verification to reveal the mechanism of colquhounia root tablet in the treatment of diabetic nephropathy. Biochem Biophys Res Commun 2024; 745:151086. [PMID: 39740397 DOI: 10.1016/j.bbrc.2024.151086] [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: 09/10/2024] [Revised: 11/09/2024] [Accepted: 11/26/2024] [Indexed: 01/02/2025]
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications of diabetes mellitus. Colquhounia Root Tablet (CRT), one of the Tripterygium wilfordii Hook F. (TwHF)-based therapeutics, has exhibited various functions in DN. However, the pharmacological mechanisms underlying its effects are still unclear. The bioactive compounds and targets of CRT were obtained from the TCMSP database, HERB-National Genomics Data Center, and SwissTargetPrediction database. The targets of DN were obtained from the DisGeNET database, Genecards database, and OMIM database. Potential therapeutic targets for CRT against DN were identified by intersecting the above targets. Protein-protein interaction (PPI), Gene ontology (GO) functional annotation, and Kyoto Encyclopedia of Genes Genomes (KEGG) enrichment analysis were performed to explore the underlying mechanism. Molecular docking assessed the binding ability between bioactive compounds and targets. Network pharmacology analysis predicted that the overlap between CRT and DN targets yielded 163 therapeutic targets. STAT3, IL1B, JUN, IL6, TNF, HIF1A, CASP3, PPARG, and BCL2 were identified as the core targets through PPI. KEGG enrichment analysis revealed that the AGEs/RAGE signaling was the prominent pathway for CRT against DN. Molecular docking indicated hypodiolide A had the lowest binding energy and the most stable binding affinity towards HIF1A. Furthermore, AGEs-induced human podocytes were treated with different concentrations of CRT to validate the predicted signaling pathway. Results showed that CRT exerted protective effects against podocyte injury, angiogenesis, and epithelial-mesenchymal transition (EMT) by regulating the AGEs/RAGE/RhoA/RCOK signaling pathway. However, in vivo studies are needed to reveal the safety and efficacy of CRT in DN.
Collapse
Affiliation(s)
- Dandan Chen
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Simeng Wang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Zhikang Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinyu Zhang
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Nannan Chong
- Department of Nephrology, Weihai Central Hospital Affiliated to Qingdao University, Weihai, Shandong, China
| | - Jianqiang Shu
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Qinglian Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Ying Xu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Chen J, Wang Z, Wang S, Lyu J, Fang Z, Qi W, Yang X, Gao G, Zhou T. Probing the familial ties between serpin members Kallistatin and PEDF: A comparative analysis review. Life Sci 2024; 362:123333. [PMID: 39719168 DOI: 10.1016/j.lfs.2024.123333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/07/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
The serine protease inhibitors (Serpins) represent a diverse protein superfamily that holds paramount significance in governing vital pathophysiological processes. Their influence on critical biological pathways renders serpins highly coveted targets for drug discovery endeavors. Among the numerous members of this family, two distinct proteins, Kallistatin (encoded by the SERPINA4 gene) and Pigment Epithelium-Derived Factor (PEDF, encoded by the SERPINF1 gene), stand out as secreted proteins that are abundantly present in peripheral blood. Kallistatin is a serine protease inhibitor that specifically inhibits human tissue kallikrein, while PEDF is a non-inhibitory member of the serine protease inhibitors superfamily (Lin et al., 2015a; Chao and Chao, 1995 [1,2]). Instead, they exhibit notable anti-angiogenic effects and play pivotal roles in the pathogenesis of metabolic disorders. Extensive research, including our own investigations, has revealed intriguing similarities as well as noteworthy differences between these two proteins. Despite their shared characteristics, the distinctive features of Kallistatin and PEDF render them unique in their respective functions and mechanisms of action. However, a comprehensive literature review comparing their similarities and differences remains elusive. Therefore, the present review aims to systematically delve into and summarize the comparable and contrasting aspects of Kallistatin and PEDF. We will delve into their expression patterns, structural features, and mechanisms of expression regulation. Furthermore, this review will delve into their physiological functions and roles in diseases, the signaling pathways they influence, and their potential clinical applications. By comparing and contrasting these two proteins, we hope to provide a comprehensive understanding of their functions and potential in biomedical research and clinical practice.
Collapse
Affiliation(s)
- Jingnan Chen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zihan Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Simin Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiayi Lyu
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhenzhen Fang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Ti Zhou
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; China Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China; Guangdong Province Key Laboratory of Diabetology, Guangzhou 510080, China.
| |
Collapse
|
3
|
Hernyák M, Tóth LI, Csiha S, Molnár Á, Lőrincz H, Paragh G, Harangi M, Sztanek F. Kallistatin as a Potential Marker of Therapeutic Response During Alpha-Lipoic Acid Treatment in Diabetic Patients with Sensorimotor Polyneuropathy. Int J Mol Sci 2024; 25:13276. [PMID: 39769041 PMCID: PMC11675709 DOI: 10.3390/ijms252413276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Diabetic sensorimotor neuropathy (DSPN) is strongly associated with the extent of cellular oxidative stress and endothelial dysfunction in type 2 diabetes (T2DM). Alpha-lipoic acid (ALA) attenuates the progression of DSPN through its antioxidant and vasculoprotective effects. Kallistatin has antioxidant and anti-inflammatory properties. We aimed to evaluate changes in kallistatin levels and markers of endothelial dysfunction in patients with T2DM and DSPN following six months of treatment with 600 mg/day of ALA. A total of 54 patients with T2DM and DSPN and 24 control patients with T2DM but without neuropathy participated in this study. The serum concentrations of kallistatin, ICAM-1, VCAM-1, oxLDL, VEGF, ADMA, and TNF-alpha were measured by an ELISA. Peripheral sensory neuropathy was assessed with neuropathy symptom questionnaires and determination of the current perception threshold. After ALA treatment, the level of kallistatin significantly decreased, as well as the levels of TNF-alpha and ADMA. Changes in kallistatin levels were positively correlated with changes in oxLDL. The improvement in DSPN symptoms following ALA treatment showed a positive correlation with changes in kallistatin, VEGF, oxLDL, and ADMA levels. Based on our results, kallistatin could represent a potential new biomarker for assessing therapeutic response during ALA treatment in patients with DSPN.
Collapse
Affiliation(s)
- Marcell Hernyák
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
- Doctoral School of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Imre Tóth
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
- Doctoral School of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary
| | - Sára Csiha
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - Ágnes Molnár
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - Hajnalka Lőrincz
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - György Paragh
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - Mariann Harangi
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
- Institute of Health Studies, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group 11003, University of Debrecen, H-4032 Debrecen, Hungary
| | - Ferenc Sztanek
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| |
Collapse
|
4
|
Gali S, Kundu A, Sharma S, Ahn MY, Puia Z, Kumar V, Kim IS, Kwak JH, Palit P, Kim HS. Therapeutic potential of bark extracts from Macaranga denticulata on renal fibrosis in streptozotocin-induced diabetic rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:911-933. [PMID: 39306745 DOI: 10.1080/15287394.2024.2394586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Macaranga denticulata (MD) bark is commonly utilized in traditional medicine for diabetes prevention and treatment. The bark extract of MD is rich in prenyl or farnesyl flavonoids and stilbenes, which possess antioxidant properties. Although data suggest the potential therapeutic benefits of the use of MD in treating diabetic nephropathy (DN), the precise mechanisms underlying MD-initiated protective effects against DN are not well understood. This study aimed to assess the renoprotective properties of MD extract by examining renofibrosis inhibition, oxidative stress, and inflammation utilizing streptozotocin-induced DN male Sprague - Dawley rats. Diabetic rats were intraperitoneally injected with streptozotocin (STZ) to induce diabetes. After 6 days, these rats were orally administered MD extract (200 mg/kg/day) or metformin (200 mg/kg/day) for 14 days. The administration of MD extract significantly lowered blood glucose levels, restored body weight, and reduced urine levels of various biomarkers associated with kidney functions. Histopathological analysis revealed protective effects in both kidneys and pancreas. Further, MD extract significantly restored abnormalities in advanced glycation end products, oxidative stress biomarkers, and proinflammatory cytokine levels in STZ-treated rats. MD extract markedly reduced renal fibrosis biomarker levels, indicating recovery from renal injury, and reversed dysregulation of sirtuins and claudin-1 in the kidneys of rats with STZ-induced diabetes. In conclusion, data demonstrated the renoprotective role of MD extract, indicating plant extract's ability to suppress oxidative stress and regulate proinflammatory pathways during pathological changes in diabetic nephropathy.
Collapse
Affiliation(s)
- Sreevarsha Gali
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Amit Kundu
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
- Department of Pharmacology, GITAM School of Pharmacy, GITAM Deemed to be University, Visakhapatnam, India
| | - Swati Sharma
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Mee-Young Ahn
- Department of Biochemistry and Health Science, Changwon National University, Changwon-si, Republic of Korea
| | - Zothan Puia
- Department of Pharmacy, Regional Institute of Paramedical & Nursing Sciences, Aizawl, India
| | - Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom Institute of Agriculture, Technology & Sciences, Allahabad, India
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Jeong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| | - Partha Palit
- Department of Pharmaceutical Sciences, Drug Discovery Research Laboratory, Assam University, Silchar, India
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, School of Pharmacy University, Suwon, Republic of Korea
| |
Collapse
|
5
|
Wen M, Sun X, Pan L, Jing S, Zhang X, Liang L, Xiao H, Liu P, Xu Z, Zhang Q, Huang H. Dihydromyricetin ameliorates diabetic renal fibrosis via regulating SphK1 to suppress the activation of NF-κB pathway. Eur J Pharmacol 2024; 978:176799. [PMID: 38945289 DOI: 10.1016/j.ejphar.2024.176799] [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: 12/12/2023] [Revised: 03/19/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
Dihydromyricetin (DHM) is a flavonoid from vine tea with broad pharmacological benefits, which improve inflammation by blocking the NF-κB pathway. A growing body of research indicates that chronic kidney inflammation is vital to the pathogenesis of diabetic renal fibrosis. Sphingosine kinase-1 (SphK1) is a key regulator of diabetic renal inflammation, which triggers the NF-κB pathway. Hence, we evaluated whether DHM regulates diabetic renal inflammatory fibrosis by acting on SphK1. Here, we demonstrated that DHM effectively suppressed the synthesis of fibrotic and inflammatory adhesion factors like ICAM-1, and VCAM-1 in streptozotocin-treated high-fat diet-induced diabetic mice and HG-induced glomerular mesangial cells (GMCs). Moreover, DHM significantly suppressed NF-κB pathway activation and reduced SphK1 activity and protein expression under diabetic conditions. Mechanistically, the results of molecular docking, molecular dynamics simulation, and cellular thermal shift assay revealed that DHM stably bound to the binding pocket of SphK1, thereby reducing sphingosine-1-phosphate content and SphK1 enzymatic activity, which ultimately inhibited NF-κB DNA binding, transcriptional activity, and nuclear translocation. In conclusion, our data suggested that DHM inhibited SphK1 phosphorylation to prevent NF-κB activation thus ameliorating diabetic renal fibrosis. This supported the clinical use and further drug development of DHM as a potential candidate for treating diabetic renal fibrosis.
Collapse
Affiliation(s)
- Min Wen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, 510801, China
| | - Xiaohong Sun
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Linjie Pan
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shujin Jing
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xuting Zhang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Liyin Liang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Peiqing Liu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhanchi Xu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Qun Zhang
- Good Clinical Practice Development, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.
| | - Heqing Huang
- Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, 510801, China.
| |
Collapse
|
6
|
Apte M, Zambre S, Pisar P, Roy B, Tupe R. Decoding the role of aldosterone in glycation-induced diabetic complications. Biochem Biophys Res Commun 2024; 721:150107. [PMID: 38781658 DOI: 10.1016/j.bbrc.2024.150107] [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: 02/17/2024] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Diabetes-mediated development of micro and macro-vascular complications is a global concern. One of the factors is hyperglycemia induced the non-enzymatic formation of advanced glycation end products (AGEs). Accumulated AGEs bind with receptor of AGEs (RAGE) causing inflammation, oxidative stress and extracellular matrix proteins (ECM) modifications responsible for fibrosis, cell damage and tissue remodeling. Moreover, during hyperglycemia, aldosterone (Aldo) secretion increases, and its interaction with mineralocorticoid receptor (MR) through genomic and non-genomic pathways leads to inflammation and fibrosis. Extensive research on individual involvement of AGEs-RAGE and Aldo-MR pathways in the development of diabetic nephropathy (DN), cardiovascular diseases (CVDs), and impaired immune system has led to the discovery of therapeutic drugs. Despite mutual repercussions, the cross-talk between AGEs-RAGE and Aldo-MR pathways remains unresolved. Hence, this review focuses on the possible interaction of Aldo and glycation in DN and CVDs, considering the clinical significance of mutual molecular targets.
Collapse
Affiliation(s)
- Mayura Apte
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Saee Zambre
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Pratiksha Pisar
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Bishnudeo Roy
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Rashmi Tupe
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| |
Collapse
|
7
|
Gao L, Xing X, Guo R, Li Q, Xu Y, Pan H, Ji P, Wang P, Yu C, Li J, An Q. Effect of Different Dietary Iron Contents on Liver Transcriptome Characteristics in Wujin Pigs. Animals (Basel) 2024; 14:2399. [PMID: 39199933 PMCID: PMC11350824 DOI: 10.3390/ani14162399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/27/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Iron is an important trace element that affects the growth and development of animals and regulates oxygen transport, hematopoiesis, and hypoxia adaptations. Wujin pig has unique hypoxic adaptability and iron homeostasis; however, the specific regulatory mechanisms have rarely been reported. This study randomly divided 18 healthy Wujin piglets into three groups: the control group, supplemented with 100 mg/kg iron (as iron glycinate); the low-iron group, no iron supplementation; and the high-iron group, supplemented with 200 mg/kg iron (as iron glycinate). The pre-feeding period was 5 days, and the formal period was 30 days. Serum was collected from empty stomachs before slaughter and at slaughter to detect changes in the serum iron metabolism parameters. Gene expression in the liver was analyzed via transcriptome analysis to determine the effects of low- and high-iron diets on transcriptome levels. Correlation analysis was performed for apparent serum parameters, and transcriptome sequencing was performed using weighted gene co-expression network analysis to reveal the key pathways underlying hypoxia regulation and iron metabolism. The main results are as follows. (1) Except for the hypoxia-inducible factor 1 (HIF-1) content (between the low- and high-iron groups), significant differences were not observed among the serum iron metabolic parameters. The serum HIF-1 content of the low-iron group was significantly higher than that of the high-iron group (p < 0.05). (2) Sequencing analysis of the liver transcriptome revealed 155 differentially expressed genes (DEGs) between the low-iron and control groups, 229 DEGs between the high-iron and control groups, and 279 DEGs between the low- and high-iron groups. Bioinformatics analysis showed that the HIF-1 and transforming growth factor-beta (TGF-β) signaling pathways were the key pathways for hypoxia regulation and iron metabolism. Four genes were selected for qPCR validation, and the results were consistent with the transcriptome sequencing data. In summary, the serum iron metabolism parameter results showed that under the influence of low- and high-iron diets, Wujin piglets maintain a steady state of physiological and biochemical indices via complex metabolic regulation of the body, which reflects their stress resistance and adaptability. The transcriptome results revealed the effects of low-iron and high-iron diets on the gene expression level in the liver and showed that the HIF-1 and TGF-β signaling pathways were key for regulating hypoxia adaptability and iron metabolism homeostasis under low-iron and high-iron diets. Moreover, HIF-1α and HEPC were the key genes. The findings provide a theoretical foundation for exploring the regulatory pathways and characteristics of iron metabolism in Wujin pigs.
Collapse
Affiliation(s)
- Lin Gao
- Yunnan Provincial Key Laboratory of Tropical and Subtropical Animal Viral Diseases, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming 650201, China;
| | - Xiaokun Xing
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Rongfu Guo
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Qihua Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Yan Xu
- Yunnan East Hunter Agriculture and Forestry Development Co., Ltd., Shuifu 657803, China;
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Peng Ji
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Ping Wang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Chuntang Yu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Jintao Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| | - Qingcong An
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.X.); (R.G.); (Q.L.); (H.P.); (P.J.); (P.W.); (C.Y.); (J.L.)
| |
Collapse
|
8
|
Vesey DA, Iyer A, Owen E, Kamato D, Johnson DW, Gobe GC, Fairlie DP, Nikolic-Paterson DJ. PAR2 activation on human tubular epithelial cells engages converging signaling pathways to induce an inflammatory and fibrotic milieu. Front Pharmacol 2024; 15:1382094. [PMID: 39005931 PMCID: PMC11239397 DOI: 10.3389/fphar.2024.1382094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/31/2024] [Indexed: 07/16/2024] Open
Abstract
Key features of chronic kidney disease (CKD) include tubulointerstitial inflammation and fibrosis. Protease activated receptor-2 (PAR2), a G-protein coupled receptor (GPCR) expressed by the kidney proximal tubular cells, induces potent proinflammatory responses in these cells. The hypothesis tested here was that PAR2 signalling can contribute to both inflammation and fibrosis in the kidney by transactivating known disease associated pathways. Using a primary cell culture model of human kidney tubular epithelial cells (HTEC), PAR2 activation induced a concentration dependent, PAR2 antagonist sensitive, secretion of TNF, CSF2, MMP-9, PAI-1 and CTGF. Transcription factors activated by the PAR2 agonist 2F, including NFκB, AP1 and Smad2, were critical for production of these cytokines. A TGF-β receptor-1 (TGF-βRI) kinase inhibitor, SB431542, and an EGFR kinase inhibitor, AG1478, ameliorated 2F induced secretion of TNF, CSF2, MMP-9, and PAI-1. Whilst an EGFR blocking antibody, cetuximab, blocked PAR2 induced EGFR and ERK phosphorylation, a TGF-βRII blocking antibody failed to influence PAR2 induced secretion of PAI-1. Notably simultaneous activation of TGF-βRII (TGF-β1) and PAR2 (2F) synergistically enhanced secretion of TNF (2.2-fold), CSF2 (4.4-fold), MMP-9 (15-fold), and PAI-1 (2.5-fold). In summary PAR2 activates critical inflammatory and fibrotic signalling pathways in human kidney tubular epithelial cells. Biased antagonists of PAR2 should be explored as a potential therapy for CKD.
Collapse
Affiliation(s)
- David A Vesey
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Abishek Iyer
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Evan Owen
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Danielle Kamato
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - David W Johnson
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - David P Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Health and Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, VIC, Australia
| |
Collapse
|
9
|
Zhong D, Chen J, Qiao R, Song C, Hao C, Zou Y, Bai M, Su W, Yang B, Sun D, Jia Z, Sun Y. Genetic or pharmacologic blockade of mPGES-2 attenuates renal lipotoxicity and diabetic kidney disease by targeting Rev-Erbα/FABP5 signaling. Cell Rep 2024; 43:114075. [PMID: 38583151 DOI: 10.1016/j.celrep.2024.114075] [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: 10/09/2023] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 04/09/2024] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common complications of diabetes, and no specific drugs are clinically available. We have previously demonstrated that inhibiting microsomal prostaglandin E synthase-2 (mPGES-2) alleviated type 2 diabetes by enhancing β cell function and promoting insulin production. However, the involvement of mPGES-2 in DKD remains unclear. Here, we aimed to analyze the association of enhanced mPGES-2 expression with impaired metabolic homeostasis of renal lipids and subsequent renal damage. Notably, global knockout or pharmacological blockage of mPGES-2 attenuated diabetic podocyte injury and tubulointerstitial fibrosis, thereby ameliorating lipid accumulation and lipotoxicity. These findings were further confirmed in podocyte- or tubule-specific mPGES-2-deficient mice. Mechanistically, mPGES-2 and Rev-Erbα competed for heme binding to regulate fatty acid binding protein 5 expression and lipid metabolism in the diabetic kidney. Our findings suggest a potential strategy for treating DKD via mPGES-2 inhibition.
Collapse
Affiliation(s)
- Dandan Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jingshuo Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Ranran Qiao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China; Public Experimental Research Center of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Chang Song
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China; Public Experimental Research Center of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Chang Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China; Public Experimental Research Center of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Yingying Zou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Mi Bai
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Wen Su
- Department of Pathophysiology, Shenzhen University, Shenzhen 518060, China; Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, Jiangsu 221002, China.
| | - Zhanjun Jia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China.
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China.
| |
Collapse
|
10
|
Peng S, Liang W, Liu Z, Ye S, Peng Z, Zhong Z, Ye Q. Hypothermic machine perfusion reduces donation after circulatory death liver ischemia-reperfusion injury through the SERPINA3-mediated PI3Kδ/Akt pathway. Hum Cell 2024; 37:420-434. [PMID: 38133876 DOI: 10.1007/s13577-023-01012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Hypothermic machine perfusion (HMP) has been demonstrated to be more effective in mitigating ischemia-reperfusion injury (IRI) of donation after circulatory death (DCD) organs than cold storage (CS), yet the underlying mechanism remains obscure. We aimed to propose a novel therapeutic approach to ameliorate IRI in DCD liver transplantation. Twelve clinical liver samples were randomly assigned to HMP or CS treatment and subsequent transcriptomics analysis was performed. By combining in vivo HMP models, we discovered that HMP attenuated inflammation, oxidative stress, and apoptosis in DCD liver through a SEPRINA3-mediated PI3Kδ/AKT signaling cascade. Moreover, in the hypoxia/reoxygenation (H/R) model of BRL-3A, overexpression of SERPINA3 mitigated H/R-induced apoptosis, while SERPINA3 knockdown exacerbated cell injury. Idelalisib (IDE) treatment also reversed the protective effect of SERPINA3 overexpression. Overall, our research provided new insights into therapeutic strategies and identified potential novel molecular targets for therapeutic intervention against DCD liver.
Collapse
Affiliation(s)
- Sheng Peng
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wenjin Liang
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Zhongzhong Liu
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Shaojun Ye
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Zibiao Zhong
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Qifa Ye
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
- Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, The 3rd Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
11
|
Zhang X, Zhang J, Ren Y, Sun R, Zhai X. Unveiling the pathogenesis and therapeutic approaches for diabetic nephropathy: insights from panvascular diseases. Front Endocrinol (Lausanne) 2024; 15:1368481. [PMID: 38455648 PMCID: PMC10918691 DOI: 10.3389/fendo.2024.1368481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/08/2024] [Indexed: 03/09/2024] Open
Abstract
Diabetic nephropathy (DN) represents a significant microvascular complication in diabetes, entailing intricate molecular pathways and mechanisms associated with cardiorenal vascular diseases. Prolonged hyperglycemia induces renal endothelial dysfunction and damage via metabolic abnormalities, inflammation, and oxidative stress, thereby compromising hemodynamics. Concurrently, fibrotic and sclerotic alterations exacerbate glomerular and tubular injuries. At a macro level, reciprocal communication between the renal microvasculature and systemic circulation establishes a pernicious cycle propelling disease progression. The current management approach emphasizes rigorous control of glycemic levels and blood pressure, with renin-angiotensin system blockade conferring renoprotection. Novel antidiabetic agents exhibit renoprotective effects, potentially mediated through endothelial modulation. Nonetheless, emerging therapies present novel avenues for enhancing patient outcomes and alleviating the disease burden. A precision-based approach, coupled with a comprehensive strategy addressing global vascular risk, will be pivotal in mitigating the cardiorenal burden associated with diabetes.
Collapse
Affiliation(s)
- Xiaoqian Zhang
- Department of Nephrology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, China
| | - Jiale Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Ren
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ranran Sun
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Zhai
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Huang X, Zhang H, Liu J, Yang X, Liu Z. Screening candidate diagnostic biomarkers for diabetic kidney disease. J Clin Lab Anal 2024; 38:e25000. [PMID: 38299750 PMCID: PMC10873681 DOI: 10.1002/jcla.25000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/25/2023] [Accepted: 12/24/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND There are big differences in treatments and prognosis between diabetic kidney disease (DKD) and non-diabetic renal disease (NDRD). However, DKD patients couldn't be diagnosed early due to lack of special biomarkers. Urine is an ideal non-invasive sample for screening DKD biomarkers. This study aims to explore DKD special biomarkers by urinary proteomics. MATERIALS AND METHODS According to the result of renal biopsy, 142 type 2 diabetes mellitus (T2DM) patients were divided into 2 groups: DKD (n = 83) and NDRD (n = 59). Ten patients were selected from each group to define urinary protein profiles by label-free quantitative proteomics. The candidate proteins were further verifyied by parallel reaction monitoring (PRM) methods (n = 40). Proteins which perform the same trend both in PRM and proteomics were verified by enzyme-linked immunosorbent assays (ELISA) with expanding the sample size (n = 82). The area under the receiver operating characteristic curve (AUC) was used to evaluate the accuracy of diagnostic biomarkers. RESULTS We identified 417 peptides in urinary proteins showing significant difference between DKD and NDRD. PRM verification identified C7, SERPINA4, IGHG1, SEMG2, PGLS, GGT1, CDH2, CDH1 was consistent with the proteomic results and p < 0.05. Three potential biomarkers for DKD, C7, SERPINA4, and gGT1, were verified by ELISA. The combinatied SERPINA4/Ucr and gGT1/Ucr (AUC = 0.758, p = 0.001) displayed higher diagnostic efficiency than C7/Ucr (AUC = 0.632, p = 0.048), SERPINA4/Ucr (AUC = 0.661, p = 0.032), and gGT1/Ucr (AUC = 0.661, p = 0.029) respectively. CONCLUSIONS The combined index SERPINA4/Ucr and gGT1/Ucr can be considered as candidate biomarkers for diabetic nephropathy after adjusting by urine creatinine.
Collapse
Affiliation(s)
- Xinying Huang
- Department of Clinical Laboratorythe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Yunnan Key Laboratory of Laboratory MedicineKunmingChina
- Yunnan Innovation Team of Clinical Laboratory and DiagnosisFirst Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Hui Zhang
- Department of Clinical Laboratorythe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Yunnan Key Laboratory of Laboratory MedicineKunmingChina
- Yunnan Innovation Team of Clinical Laboratory and DiagnosisFirst Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Jihong Liu
- Department of Clinical Laboratorythe Third People's Hospital of KunmingKunmingChina
| | - Xuejiao Yang
- Department of Clinical Laboratorythe People's Hospital of ChuXiong Yi Autonomous PrefectureChuXiongChina
| | - Zijie Liu
- Department of Clinical Laboratorythe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
- Yunnan Key Laboratory of Laboratory MedicineKunmingChina
- Yunnan Innovation Team of Clinical Laboratory and DiagnosisFirst Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
13
|
Gu MJ, Lee HW, Yoo G, Kim D, Kim Y, Choi IW, Cha YS, Ha SK. Hippophae rhamnoides L. leaf extracts alleviate diabetic nephropathy via attenuation of advanced glycation end product-induced oxidative stress in db/db mice. Food Funct 2023; 14:8396-8408. [PMID: 37614189 DOI: 10.1039/d3fo01364b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Diabetes mellitus leads to chronic complications, such as nephropathy. Diabetic complications are closely related to advanced glycation end products (AGEs). Excessive formation and accumulation of AGEs in diabetic renal diseases lead to excessive oxidative stress, resulting in chronic renal failure. The leaves of Hippophae rhamnoides L. (sea buckthorn leaves; SBL) show biological benefits, including antioxidant effects. This study aimed to evaluate the effect of SBL on kidney damage in db/db mice. The SBL extract was orally administered at 100 and 200 mg kg-1 for 12 weeks to db/db mice. Histological changes and the urine albumin/creatinine ratio were relieved, and the accumulation of AGEs in kidney glomeruli decreased following SBL treatment. Moreover, the SBL extract reduced the expression of AGEs, the receptor for AGEs, and NADPH oxidase 4, but upregulated glyoxalase 1 in the diabetic renal tissue. Urinary excretion levels and expression of 8-hydroxy-2'-deoxyguanosine as a biomarker of oxidative stress decreased after SBL treatment in the renal tissue. Furthermore, SBL attenuated oxidative stress in diabetic kidneys by reducing AGE accumulation, thereby ameliorating renal damage. Therefore, from these results, we infer that the SBL extract can act as a potential therapeutic agent for diabetic renal complications caused by AGEs.
Collapse
Affiliation(s)
- Min Ji Gu
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- Department of Food Science and Human Nutrition (Human Ecology), Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Hee-Weon Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Guijae Yoo
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Donghwan Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Yoonsook Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - In-Wook Choi
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition (Human Ecology), Jeonbuk National University, Jeonju 54896, Republic of Korea
- K-Food Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Sang Keun Ha
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| |
Collapse
|
14
|
Shen G, Li Y, Zeng Y, Hong F, Zhang J, Wang Y, Zhang C, Xiang W, Wang J, Fang Z, Qi W, Yang X, Gao G, Zhou T. Kallistatin Deficiency Induces the Oxidative Stress-Related Epithelial-Mesenchymal Transition of Retinal Pigment Epithelial Cells: A Novel Protagonist in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2023; 64:15. [PMID: 37682567 PMCID: PMC10500364 DOI: 10.1167/iovs.64.12.15] [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: 04/03/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023] Open
Abstract
Purpose Retinal pigment epithelium (RPE) dysfunction induced by oxidative stress-related epithelial-mesenchymal transition (EMT) of RPE is the primary underlying mechanism of age-related macular degeneration (AMD). Kallistatin (KAL) is a secreted protein with an antioxidative stress effect. However, the relationship between KAL and EMT in RPE has not been determined. Therefore we aimed to explore the impact and mechanism of KAL in oxidative stress-induced EMT of RPE. Methods Sodium iodate (SI) was injected intraperitoneally to construct the AMD rat model and investigate the changes in RPE morphology and KAL expression. KAL knockout rats and KAL transgenic mice were used to explain the effects of KAL on EMT and oxidative stress. In addition, Snail overexpressed adenovirus and si-RNA transfected ARPE19 cells to verify the involvement of Snail in mediating KAL-suppressed EMT of RPE. Results AMD rats induced by SI expressed less KAL in the retina, and KAL knockout rats showed RPE dysfunction spontaneously where EMT and reactive oxygen species (ROS) production increased in RPE. In contrast, KAL overexpression attenuated EMT and ROS levels in RPE, even in TGF-β treatment. Mechanistically, Snail reversed the beneficial effect of KAL on EMT and ROS reduction. Moreover, KAL ameliorated SI-induced AMD-like pathological changes. Conclusions Our findings demonstrated that KAL inhibits oxidative stress-induced EMT by downregulating the transcription factor Snail. Herein, KAL knockout rats may be an appropriate animal model for observing spontaneous RPE dysfunction for AMD-like retinopathy, and KAL may represent a novel therapeutic target for treating dry AMD.
Collapse
Affiliation(s)
- Gang Shen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanmei Li
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yongcheng Zeng
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Fuyan Hong
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jing Zhang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yan Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Chengwei Zhang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wei Xiang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jinhong Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zhenzhen Fang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-Sen University, Guangzhou, China
| | - Ti Zhou
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- China Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
15
|
Wang X, Huang X, Gao P, Ren Y, Li X, Diao Y. Kallistatin attenuates inflammatory response in rheumatoid arthritis via the NF-κB signaling pathway. Eur J Pharmacol 2023; 943:175530. [PMID: 36690053 DOI: 10.1016/j.ejphar.2023.175530] [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: 10/18/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Cartilage degeneration and inflammation are important features of rheumatoid arthritis (RA). Chondrocyte inflammation and apoptosis have been increasingly demonstrated to be related to cartilage decomposition. In this study, we analyzed the protective role of kallistatin against RA and its associated mechanisms. We obtained in vitro and in vivo RA models using IL-1β and heat-inactivated Mycobacterium tuberculosis, respectively. Our results showed that kallistatin mitigated IL-1β-mediated chondrocyte apoptosis and inhibited the synthesis of ECM-degrading generation, like matrix metalloproteinase (MMP)-3/13 and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4/5, in IL-1β-mediated chondrocytes. Furthermore, kallistatin markedly suppressed IL-1β-mediated inflammation while decreasing the levels of inflammatory factors and mediators via the NF-κB pathway. Daily administration of kallistatin reduced the expression levels of PGE2, TNF-α, IL-1β, and IL-6. Histochemical analysis revealed that the kallistatin-treated rats exhibited reduced RA severity compared with control mice. In summary, kallistatin suppressed IL-1β-mediated inflammation in chondrocytes via the NF-κB pathway. Administration of kallistatin remarkably inhibited RA development, accompanied by reduced inflammation and apoptosis. Therefore, kallistatin administration can be used as a candidate therapeutic strategy for RA.
Collapse
Affiliation(s)
- Xiao Wang
- School of Medicine, Huaqiao University, Quanzhou, 362021, China
| | - Xiaoping Huang
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, 326000, China
| | - Pingzhang Gao
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, 326000, China
| | - Yanxuan Ren
- School of Medicine, Huaqiao University, Quanzhou, 362021, China
| | - Xiaokun Li
- School of Medicine, Huaqiao University, Quanzhou, 362021, China
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou, 362021, China.
| |
Collapse
|
16
|
Yiu W, Lok S, Xue R, Chen J, Lai K, Lan H, Tang S. The long noncoding RNA Meg3 mediates TLR4-induced inflammation in experimental obstructive nephropathy. Clin Sci (Lond) 2023; 137:317-331. [PMID: 36705251 PMCID: PMC9977690 DOI: 10.1042/cs20220537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/28/2023]
Abstract
Kidney inflammation contributes to the progression of chronic kidney disease (CKD). Modulation of Toll-like receptor 4 (TLR4) signaling is a potential therapeutic strategy for this pathology, but the regulatory mechanisms of TLR4 signaling in kidney tubular inflammation remains unclear. Here, we demonstrated that tubule-specific deletion of TLR4 in mice conferred protection against obstruction-induced kidney injury, with reduction in inflammatory cytokine production, macrophage infiltration and kidney fibrosis. Transcriptome analysis revealed a marked down-regulation of long noncoding RNA (lncRNA) Meg3 in the obstructed kidney from tubule-specific TLR4 knockout mice compared with wild-type control. Meg3 was also induced by lipopolysaccharide in tubular epithelial cells via a p53-dependent signaling pathway. Silencing of Meg3 suppressed LPS-induced cytokine production of CCL-2 and CXCL-2 and the activation of p38 MAPK pathway in vitro and ameliorated kidney fibrosis in mice with obstructive nephropathy. Together, these findings identify a proinflammatory role of lncRNA Meg3 in CKD and suggest a novel regulatory pathway in TLR4-driven inflammatory responses in tubular epithelial cells.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Sarah W.Y. Lok
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Rui Xue
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Jiaoyi Chen
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kar Neng Lai
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Hui Yao Lan
- Department of Medicine and Therapeutics and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Sydney C.W. Tang
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
- Correspondence: Sydney C.W. Tang ()
| |
Collapse
|
17
|
Tsai MT, Yang RB, Ou SM, Tseng WC, Lee KH, Yang CY, Chang FP, Tarng DC. Plasma Galectin-9 Is a Useful Biomarker for Predicting Renal Function in Patients Undergoing Native Kidney Biopsy. Arch Pathol Lab Med 2023; 147:167-176. [PMID: 35687787 DOI: 10.5858/arpa.2021-0466-oa] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 02/05/2023]
Abstract
CONTEXT.— Galectin-9 reduces tissue damage in certain immune-mediated glomerular diseases. However, its role in structural and functional renal changes in patients with varying types of chronic kidney disease (CKD) is less clear. OBJECTIVE.— To investigate the association between plasma galectin-9 levels, proteinuria, tubulointerstitial lesions, and renal function in different CKD stages. DESIGN.— We measured plasma galectin-9 levels in 243 patients undergoing renal biopsy for determining the CKD etiology. mRNA and protein expression levels of intrarenal galectin-9 were assessed by quantitative real-time polymerase chain reaction and immunostaining. Relationships between plasma galectin-9, clinical characteristics, and tubulointerstitial damage were analyzed with logistic regression. We investigated galectin-9 expression patterns in vitro in murine J774 macrophages treated with differing stimuli. RESULTS.— To analyze the relationship between galectin-9 and clinical features, we divided the patients into 2 groups according to median plasma galectin-9 levels. The high galectin-9 group tended to be older and to have decreased renal function, higher proteinuria, and greater interstitial fibrosis. After multivariable adjustment, elevated plasma galectin-9 levels were independently associated with stage 3b or higher CKD. An analysis of gene expression in the tubulointerstitial compartment in the biopsy samples showed a significant positive correlation between intrarenal galectin-9 mRNA expression and plasma galectin-9 levels. Immunohistochemistry confirmed increased galectin-9 expression in the renal interstitium of patients with advanced CKD, and most galectin-9-positive cells were macrophages, as determined by double-immunofluorescence staining. In vitro experiments showed that galectin-9 expression in macrophages was significantly increased after interferon-γ stimulation. CONCLUSIONS.— Our findings suggest that plasma galectin-9 is a good biomarker for diagnosing advanced CKD.
Collapse
Affiliation(s)
- Ming-Tsun Tsai
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan.,Tsai and R-B Yang contributed equally to this manuscript
| | - Ruey-Bing Yang
- From the Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (R-B Yang).,Tsai and R-B Yang contributed equally to this manuscript
| | - Shuo-Ming Ou
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Cheng Tseng
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuo-Hua Lee
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Yu Yang
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fu-Pang Chang
- From the Department of Pathology and Laboratory Medicine (Chang), Taipei Veterans General Hospital, Taipei, Taiwan
| | - Der-Cherng Tarng
- From the Division of Nephrology, Department of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), Taipei Veterans General Hospital, Taipei, Taiwan.,From the Institute of Clinical Medicine, School of Medicine (Tsai, Ou, Tseng, Lee, C-Y Yang, Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan.,From the Department and Institute of Physiology (Tarng), National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
18
|
Fan Z, Gao Y, Jiang N, Zhang F, Liu S, Li Q. Immune-related SERPINA3 as a biomarker involved in diabetic nephropathy renal tubular injury. Front Immunol 2022; 13:979995. [DOI: 10.3389/fimmu.2022.979995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease and has become a serious medical issue globally. Although it is known to be associated with glomerular injury, tubular injury has been found to participate in DN in recent years. However, mechanisms of diabetic renal tubular injury remain unclear. Here, we investigated the differentially expressed genes in the renal tubules of patients with DN by analyzing three RNA-seq datasets downloaded from the Gene Expression Omnibus database. Gene set enrichment analysis and weighted gene co-expression network analysis showed that DN is highly correlated with the immune system. The immune-related gene SERPINA3 was screened out with lasso regression and Kaplan–Meier survival analyses. Considering that SERPINA3 is an inhibitor of mast cell chymase, we examined the expression level of SERPINA3 and chymase in human renal tubular biopsies and found that SERPINA3 was upregulated in DN tubules, which is consistent with the results of the differential expression analysis. Besides, the infiltration and degranulation rates of mast cells are augmented in DN. By summarizing the biological function of SERPINA3, chymase, and mast cells in DN based on our results and those of previous studies, we speculated that SERPINA3 is a protective immune-related molecule that prevents renal tubular injury by inhibiting the proliferation and activation of mast cells and downregulating the activity of chymase.
Collapse
|
19
|
Huang X, Li B, Hu J, Liu Z, Li D, Chen Z, Huang H, Chen Y, Guo X, Cui Y, Huang Q. Advanced glycation endproducts mediate chronic kidney injury with characteristic patterns in different stages. Front Physiol 2022; 13:977247. [PMID: 36160865 PMCID: PMC9500449 DOI: 10.3389/fphys.2022.977247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022] Open
Abstract
Advanced glycation endproducts (AGEs) have been confirmed to play a causative role in the development of diabetic nephropathy (DN). In this study, we revealed that AGE-induced kidney injury with characteristic patterns in different stages and moesin phosphorylation plays a role in these processes. In WT mice treated with AGE-modified bovine serum albumin (AGE-BSA), distinct abnormal angiogenesis in Bowman’s capsule of the kidney emerged early after 1 m under AGE-BSA stimulation, while these neovessels became rare after 6 m. AGE-BSA also induced glomerular hypertrophy and mesangial expansion at 1 m but glomerular atrophy and fibrosis at 6 m. Electron microscopy imaging demonstrated the damage of foot process integrity in podocytes and the uneven thickening of the glomerular basement membrane in the AGE-BSA-treated group, which was more significant after 6 m of AGE-BSA treatment than 1 m. The kidney dysfunction appeared along with these AGE-induced morphological changes. However, these AGE-BSA-induced pathological changes were significantly attenuated in RAGE-knockout mice. Moreover, moesin phosphorylation was accompanied by AGE-BSA-induced alterations and moesin deficiency in mice attenuated by AGE-BSA-induced fibrosis. The investigation on glomerular endothelial cells (GECs) also confirmed that the phosphorylation of moesin T558 is critical in AGE-induced tube formation. Overall, this study suggests that AGEs mediate kidney injury with characteristic patterns by binding with RAGE and inducing moesin phosphorylation.
Collapse
Affiliation(s)
- Xiaoxia Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bingyu Li
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiaqing Hu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuanhua Liu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dongping Li
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenfeng Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hang Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanjia Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yun Cui
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Guangzhou, China
- *Correspondence: Yun Cui, ; Qiaobing Huang,
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Yun Cui, ; Qiaobing Huang,
| |
Collapse
|
20
|
Harkin C, Smith KW, MacKay CL, Moore T, Brockbank S, Ruddock M, Cobice DF. Spatial localization of β-unsaturated aldehyde markers in murine diabetic kidney tissue by mass spectrometry imaging. Anal Bioanal Chem 2022; 414:6657-6670. [PMID: 35881173 PMCID: PMC9411223 DOI: 10.1007/s00216-022-04229-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022]
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. Limitations in current diagnosis and screening methods have sparked a search for more specific and conclusive biomarkers. Hyperglycemic conditions generate a plethora of harmful molecules in circulation and within tissues. Oxidative stress generates reactive α-dicarbonyls and β-unsaturated hydroxyhexenals, which react with proteins to form advanced glycation end products. Mass spectrometry imaging (MSI) enables the detection and spatial localization of molecules in biological tissue sections. Here, for the first time, the localization and semiquantitative analysis of “reactive aldehydes” (RAs) 4-hydroxyhexenal (4-HHE), 4-hydroxynonenal (4-HNE), and 4-oxo-2-nonenal (4-ONE) in the kidney tissues of a diabetic mouse model is presented. Ionization efficiency was enhanced through on-tissue chemical derivatization (OTCD) using Girard’s reagent T (GT), forming positively charged hydrazone derivatives. MSI analysis was performed using matrix-assisted laser desorption ionization (MALDI) coupled with Fourier-transform ion cyclotron resonance mass spectrometry (FT-ICR). RA levels were elevated in diabetic kidney tissues compared to lean controls and localized throughout the kidney sections at a spatial resolution of 100 µm. This was confirmed by liquid extraction surface analysis–MSI (LESA-MSI) and liquid chromatography–mass spectrometry (LC–MS). This method identified β-unsaturated aldehydes as “potential” biomarkers of DN and demonstrated the capability of OTCD-MSI for detection and localization of poorly ionizable molecules by adapting existing chemical derivatization methods. Untargeted exploratory distribution analysis of some precursor lipids was also assessed using MALDI-FT-ICR-MSI.
Collapse
Affiliation(s)
- Carla Harkin
- Mass Spectrometry Centre, Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Karl W Smith
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, 32310-4005, USA.,Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - C Logan MacKay
- Scottish Instrumentation and Research Centre for Advanced Mass Spectrometry (SIRCAMS), EastChem School of Chemistry, University of Edinburgh, Edinburgh, Scotland, UK
| | - Tara Moore
- Genomic Medicine, Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | | | - Mark Ruddock
- Randox Laboratories Ltd, 55 The Diamond Rd, Crumlin, UK
| | - Diego F Cobice
- Mass Spectrometry Centre, Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
21
|
Exogenous pancreatic kininogenase protects against tacrolimus-induced renal injury by inhibiting PI3K/AKT signaling: The role of bradykinin receptors. Int Immunopharmacol 2022; 105:108547. [DOI: 10.1016/j.intimp.2022.108547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/20/2022]
|
22
|
El-Hefnawy SM, Kasemy ZA, Eid HA, Elmadbouh I, Mostafa RG, Omar TA, Kasem HE, Ghonaim EM, Ghonaim MM, Saleh AA. Potential impact of serpin peptidase inhibitor clade (A) member 4 SERPINA4 (rs2093266) and SERPINA5 (rs1955656) genetic variants on COVID-19 induced acute kidney injury. Meta Gene 2022:101023. [PMID: 35291551 PMCID: PMC8915573 DOI: 10.1016/j.mgene.2022.101023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 11/24/2022] Open
Abstract
Background SARS-CoV-2 has a number of targets, including the kidneys. Acute Kidney Injury (AKI) might develop in up to a quarter of SARS-CoV-2 patients. In the clinical environment, AKI is linked to a high rate of death and leads to the progression of AKI to chronic renal disease. Aim We aimed to investigate rs2093266 and rs1955656 polymorphisms in SERPINA4 and SERPINA5 genes, respectively, as risk factors for COVID-19 induced AKI. Subjects and methods A case-control study included 227 participants who were divided into three groups: 81 healthy volunteers who served as controls, 76 COVID-19 patients without AKI and 70 COVID -19 patients with AKI. The TaqMan assay was used for genotyping the SERPINA4 (rs2093266) and SERPINA5 (rs1955656) polymorphisms by real-time PCR technique. Results Lymphocytes and eGFR showed a significantly decreasing trend across the three studied groups, while CRP, d-Dimer, ferritin, creatinine, KIM-1and NGAL showed a significantly increasing trend across the three studied groups (P < 0.001). Rs2093266 (AG and AA) genotypes were significant risk factors among non-AKI and AKI groups in comparison to controls. Rs1955656 (AG and AA) were significant risk factors among the AKI group, while AA was the only significant risk factor among the non-AKI group. Recessive, dominant, co-dominant, and over-dominant models for genotype combinations were demonstrated. The GG v AA, GG + AG v AA, and GG v AG + AA models of the rs2093266 were all significant predictors of AKI, whilst only the GG v AA model of the rs1955656 SNP was a significant predictor. The logistic regression model was statistically significant, χ2 = 56.48, p < 0.001. AKI was associated with progressed age (OR = 0.95, 95% CI: 0.91–0.98, p = 0.006), suffering from chronic diseases (OR = 3.25, 95% CI: 1.31–8.01, p = 0.010), increased BMI (OR = 0.89, 95% CI: 0.81–0.98, p = 0.018), immunosuppressive (OR = 4.61, 95% CI: 1.24–17.16, p = 0.022) and rs2093266 (AG + AA) (OR = 3.0, 95% CI: 1.11–8.10, p = 0.030). Conclusion Single nucleotide polymorphisms (rs2093266) at SERPINA4 gene and (rs1955656) at SERPINA5 gene were strongly linked to the development of AKI in COVID-19 patients.
Collapse
|
23
|
Hu L, Ding M, He W. Emerging Therapeutic Strategies for Attenuating Tubular EMT and Kidney Fibrosis by Targeting Wnt/β-Catenin Signaling. Front Pharmacol 2022; 12:830340. [PMID: 35082683 PMCID: PMC8784548 DOI: 10.3389/fphar.2021.830340] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 12/25/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is defined as a process in which differentiated epithelial cells undergo phenotypic transformation into myofibroblasts capable of producing extracellular matrix, and is generally regarded as an integral part of fibrogenesis after tissue injury. Although there is evidence that the complete EMT of tubular epithelial cells (TECs) is not a major contributor to interstitial myofibroblasts in kidney fibrosis, the partial EMT, a status that damaged TECs remain inside tubules, and co-express both epithelial and mesenchymal markers, has been demonstrated to be a crucial stage for intensifying fibrogenesis in the interstitium. The process of tubular EMT is governed by multiple intracellular pathways, among which Wnt/β-catenin signaling is considered to be essential mainly because it controls the transcriptome associated with EMT, making it a potential therapeutic target against kidney fibrosis. A growing body of data suggest that reducing the hyperactivity of Wnt/β-catenin by natural compounds, specific inhibitors, or manipulation of genes expression attenuates tubular EMT, and interstitial fibrogenesis in the TECs cultured under profibrotic environments and in animal models of kidney fibrosis. These emerging therapeutic strategies in basic researches may provide beneficial ideas for clinical prevention and treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Lichao Hu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Mengyuan Ding
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Weichun He
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Zhang Z, Hu Y, Liu W, Zhang X, Wang R, Li H, Sun D, Fang J. Yishen Capsule Alleviated Symptoms of Diabetic Nephropathy via NOD-like Receptor Signaling Pathway. Diabetes Metab Syndr Obes 2022; 15:2183-2195. [PMID: 35923253 PMCID: PMC9339947 DOI: 10.2147/dmso.s368867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To explore the mechanism of Yishen capsule against diabetic nephropathy (DN) based on the analysis of transcriptomics. MATERIAL AND METHODS SD rats (Male, SPF grade) were randomly divided into four groups, the normal group, the DN group, the Yishen capsule group and the resveratrol group. Urine and renal tissue samples were collected after feeding with physiological saline and above drugs for 8 weeks. 24-hour urine microalbumin protein was detected by ELISA. HE staining and PAS staining were performed on renal tissues. Differential gene expression in renal tissues was analyzed by transcriptome sequencing. The differentially expressed genes were analyzed by GO enrichment and KEGG enrichment, and verified by RT-PCR and immunohistochemistry staining. RESULTS The level of 24-hour urinary microalbumin in DN group was increased, while Yishen capsule treatment reversed the increasement of urinary microalbumin. Mesangial cell proliferation, matrix accumulation, edema and vacuolar degeneration of renal tubular epithelial cells and glycogen accumulation were observed in DN group. However, pathological phenotypes mentioned above were alleviated after Yisen capsule administration. This result indicates that Yishen capsule reversed pathological phenotypes of DN in rats. The expression of 261 genes were changed in Yishen capsule group compared with DN group. GO enrichment analysis and KEGG pathway analysis showed that these genes were implicated in pathways, including mineral absorption, adipocytokine signaling pathway, fatty acid biosynthesis, thyroid hormone synthesis, renin-angiotensin system, and NOD-like receptor signaling pathway. Based on previous reported study, the expression of key factors in NOD-like receptor signaling pathway was verified. RT-PCR and immunohistochemistry staining showed that the expression of NLRP3, Caspase-1 and IL-1β in renal tissues of DN group were increased (P < 0.05), which were decreased in Yishen capsule group (P < 0.05). CONCLUSION Yishen capsule reduced microalbuminuria and alleviated pathological changes in DN rats, which may be achieved by regulating NOD-like receptor signaling pathway.
Collapse
Affiliation(s)
- Ziyuan Zhang
- Shanxi Medical University, Taiyuan, People’s Republic of China
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yaling Hu
- Shanxi Medical University, Taiyuan, People’s Republic of China
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Wenyuan Liu
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xiaodong Zhang
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Ruihua Wang
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Hui Li
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Dalin Sun
- Shanxi Medical University, Taiyuan, People’s Republic of China
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jingai Fang
- Department of Nephrology, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Correspondence: Jingai Fang, Department of Nephrology, First Hospital of Shanxi Medical University, 85 Jiefangnan Road, Taiyuan, 030001, People’s Republic of China, Email
| |
Collapse
|
25
|
Wen S, Deng F, Li L, Xu L, Li X, Fan Q. VX-765 ameliorates renal injury and fibrosis in diabetes by regulating caspase-1-mediated pyroptosis and inflammation. J Diabetes Investig 2022; 13:22-33. [PMID: 34494385 PMCID: PMC8756311 DOI: 10.1111/jdi.13660] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION As a lytic inflammatory cell death, pyroptosis has been recently described but has not been unequivocally elucidated in diabetic nephropathy (DN). VX-765 is a safe and effective inhibitor of caspase-1, that was well tolerated in a phase II clinical trial in patients with epilepsy, but its application in DN is still undefined. MATERIALS AND METHODS Immunoblot, co-immunoprecipitation, confocal microscope and flow cytometry were used to analyze the effects of glucose on pyroptosis in renal tubular epithelia (HK-2). In vitro, selective caspase-1 inhibitors VX-765 and Z-YVAD-FMK were administered. Pyroptosis and fibrogenesis were determined by immunoblot, ELISA, cytotoxicity assay and flow cytometry. In vivo, diabetic mice were administered with 100 mg/kg VX-765. Renal function, pathological changes, and the expressions of NLRC4, GSDMD, IL-1β, collagen I, fibronectin and CD45 in renal cortex were evaluated. RESULTS We identified NLRC4 as a sensor for caspase-1 activation. Moreover, we provided morphological and molecular evidence for pyroptosis in glucose-stressed tubular cells, including ballooned cell membrane, caspase-1 immunoreactivity, GSDMD cleavage, and the release of inflammatory cytokine and cellular contents. All these effects were prevented by treatment with VX-765 or Z-YVAD-FMK, confirming that caspase-1 effectively regulates the occurrence of pyroptosis in HK-2 cells. In vivo, treatment of diabetic animals with VX-765 ameliorated renal function, suppressed inflammatory cell infiltration and pyroptosis-associated protein expression, and mitigated tubulointerstitial fibrosis. CONCLUSIONS This work revealed that caspase-1-mediated pyroptosis drives renal inflammation and fibrosis in diabetes. Our results are the first demonstration of VX-765 representing a promising therapeutic opportunity for alleviating the progression of DN.
Collapse
Affiliation(s)
- Si Wen
- Department of NephrologyFirst Hospital of China Medical UniversityShenyangChina
- Department of NephrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Fei Deng
- Department of UrologySecond Xiangya Hospital of Central South UniversityChangshaChina
| | - Lulu Li
- Department of NephrologyFirst Hospital of China Medical UniversityShenyangChina
| | - Li Xu
- Department of NephrologyFirst Hospital of China Medical UniversityShenyangChina
- Department of Laboratory MedicineFirst Hospital of China Medical UniversityShenyangChina
| | - Xin Li
- Department of NephrologyFirst Hospital of China Medical UniversityShenyangChina
| | - Qiuling Fan
- Department of NephrologyFirst Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
26
|
Spatial-resolved metabolomics reveals tissue-specific metabolic reprogramming in diabetic nephropathy by using mass spectrometry imaging. Acta Pharm Sin B 2021; 11:3665-3677. [PMID: 34900545 PMCID: PMC8642449 DOI: 10.1016/j.apsb.2021.05.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
Detailed knowledge on tissue-specific metabolic reprogramming in diabetic nephropathy (DN) is vital for more accurate understanding the molecular pathological signature and developing novel therapeutic strategies. In the present study, a spatial-resolved metabolomics approach based on air flow-assisted desorption electrospray ionization (AFADESI) and matrix-assisted laser desorption ionization (MALDI) integrated mass spectrometry imaging (MSI) was proposed to investigate tissue-specific metabolic alterations in the kidneys of high-fat diet-fed and streptozotocin (STZ)-treated DN rats and the therapeutic effect of astragaloside IV, a potential anti-diabetic drug, against DN. As a result, a wide range of functional metabolites including sugars, amino acids, nucleotides and their derivatives, fatty acids, phospholipids, sphingolipids, glycerides, carnitine and its derivatives, vitamins, peptides, and metal ions associated with DN were identified and their unique distribution patterns in the rat kidney were visualized with high chemical specificity and high spatial resolution. These region-specific metabolic disturbances were ameliorated by repeated oral administration of astragaloside IV (100 mg/kg) for 12 weeks. This study provided more comprehensive and detailed information about the tissue-specific metabolic reprogramming and molecular pathological signature in the kidney of diabetic rats. These findings highlighted the promising potential of AFADESI and MALDI integrated MSI based metabolomics approach for application in metabolic kidney diseases.
Collapse
Key Words
- ADP, adenosine diphosphate
- AFADESI, air flow-assisted desorption electrospray ionization
- AGEs, advanced glycation end products
- AMP, adenosine monophosphate
- AMPK, adenosine monophosphate activated protein kinase
- AST, astragaloside IV
- ATP, adenosine triphosphate
- Astragaloside IV
- BUN, blood urea nitrogen
- CL, cardiolipin
- Cre, creatinine
- DAG, diacylglycerol
- DESI, desorption electrospray ionization
- DM, diabetes mellitus
- DN, diabetic nephropathy
- DPA, docosapentaenoic acid
- Diabetic nephropathy
- ESKD, end-stage kidney disease
- FBG, fasting blood glucose
- GLU, glucose
- GMP, guanosine monophosphate
- GSH, glutathione
- H&E, hematoxylin and eosin
- HPLC, high-performance liquid chromatography
- HbA1c, glycosylated hemoglobin
- LysoPC, lysophosphatidylcholine
- LysoPG, lysophosphatidylglycerol
- MALDI, matrix-assisted laser desorption ionization
- MS, mass spectrometry
- MSI, mass spectrometry imaging
- Mass spectrometry imaging
- Metabolic reprogramming
- NMR, nuclear magnetic resonance
- Na-CMC, sodium carboxymethyl cellulose
- PA, phosphatidic acid
- PC, phosphatidylcholine
- PE, phosphatidylethanolamine
- PG, phosphatidylglycerol
- PPP, pentose phosphate pathway
- PS, phosphatidylserine
- PUFA, polyunsaturated fatty acids
- ROI, regions of interest
- ROS, reactive oxygen species
- SDH, succinate dehydrogenase
- SGLTs, sodium-glucose cotransporters
- SM, sphingomyelin
- STZ, streptozotocin
- Spatial-resolved metabolomics
- TCA, tricarboxylic acid
- TCHO, total cholesterol
- TG, triglyceride
- UMP, uridine monophosphate
- VIP, variable importance in projection
- p-AMPK, phosphorylated adenosine monophosphate activated protein kinase
Collapse
|
27
|
Chang DY, Li XQ, Chen M, Zhao MH. Dapagliflozin Ameliorates Diabetic Kidney Disease via Upregulating Crry and Alleviating Complement Over-activation in db/db Mice. Front Pharmacol 2021; 12:729334. [PMID: 34712135 PMCID: PMC8546210 DOI: 10.3389/fphar.2021.729334] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/29/2021] [Indexed: 12/30/2022] Open
Abstract
Sodium-glucose cotransporter 2(SGLT2) inhibitors show prominent renal protective effect in diabetic kidney disease (DKD), anti-inflammatory effect being one of its key mechanisms. Over-activation of the complement system, a crucial part of innate immunity, plays an important role in DKD. We aimed to investigate the effect of SGLT2 inhibitors on alleviating complement over-activation in DKD. Db/db mice were randomly divided into two groups, with 7 mice in each group treated with dapagliflozin and vehicle respectively, and 7 mice in m/m mice group. Laboratory and renal pathological parameters were evaluated. Mouse proximal tubular epithelial cells (MPTECs) were cultured and treated with high glucose. Dapagliflozin and dimethyloxallyl glycine (DMOG) were added as conditional treatment. Dapagliflozin-treated db/db mice showed significantly lower urinary albumin than vehicle-treated ones. Besides typical glomerular and tubulointerstitial injury, both C3b and membrane attack complex (MAC) depositions were significantly attenuated in dapagliflozin-treated db/db mice. The expression of complement receptor type 1-related protein y (Crry), a key complement regulator which inhibits complement over-activation, was significantly upregulated by dapagliflozin. Dapagliflozin-mediated Crry upregulation was associated with inhibition of HIF-1α accumulation under high glucose. When HIF-1α expression was stabilized by DMOG, the protective effect of dapagliflozin via upregulating Crry was blocked. In conclusion, dapagliflozin could attenuate complement over-activation in diabetic mice via upregulating Crry, which is associated with the suppression of HIF-1α accumulation in MPTECs.
Collapse
Affiliation(s)
- Dong-Yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Qian Li
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Krishna SM, Li J, Wang Y, Moran CS, Trollope A, Huynh P, Jose R, Biros E, Ma J, Golledge J. Kallistatin limits abdominal aortic aneurysm by attenuating generation of reactive oxygen species and apoptosis. Sci Rep 2021; 11:17451. [PMID: 34465809 PMCID: PMC8408144 DOI: 10.1038/s41598-021-97042-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/20/2021] [Indexed: 11/09/2022] Open
Abstract
Inflammation, vascular smooth muscle cell apoptosis and oxidative stress are believed to play important roles in abdominal aortic aneurysm (AAA) pathogenesis. Human kallistatin (KAL; gene SERPINA4) is a serine proteinase inhibitor previously shown to inhibit inflammation, apoptosis and oxidative stress. The aim of this study was to investigate the role of KAL in AAA through studies in experimental mouse models and patients. Serum KAL concentration was negatively associated with the diagnosis and growth of human AAA. Transgenic overexpression of the human KAL gene (KS-Tg) or administration of recombinant human KAL (rhKAL) inhibited AAA in the calcium phosphate (CaPO4) and subcutaneous angiotensin II (AngII) infusion mouse models. Upregulation of KAL in both models resulted in reduction in the severity of aortic elastin degradation, reduced markers of oxidative stress and less vascular smooth muscle apoptosis within the aorta. Administration of rhKAL to vascular smooth muscle cells incubated in the presence of AngII or in human AAA thrombus-conditioned media reduced apoptosis and downregulated markers of oxidative stress. These effects of KAL were associated with upregulation of Sirtuin 1 activity within the aortas of both KS-Tg mice and rodents receiving rhKAL. These results suggest KAL-Sirtuin 1 signalling limits aortic wall remodelling and aneurysm development through reductions in oxidative stress and vascular smooth muscle cell apoptosis. Upregulating KAL may be a novel therapeutic strategy for AAA.
Collapse
Affiliation(s)
- Smriti Murali Krishna
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - Jiaze Li
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - Yutang Wang
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Horsham, VIC, Australia
| | - Corey S Moran
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - Alexandra Trollope
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,Division of Anatomy, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | - Pacific Huynh
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - Roby Jose
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - Erik Biros
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia
| | - Jianxing Ma
- Department of Physiology, Health Sciences Centre, University of Oklahoma, Oklahoma City, OK, 73104, USA
| | - Jonathan Golledge
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia. .,Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD, Australia.
| |
Collapse
|
29
|
Li T, Yang Y, Wang X, Dai W, Zhang L, Piao C. Flavonoids derived from buckwheat hull can break advanced glycation end-products and improve diabetic nephropathy. Food Funct 2021; 12:7161-7170. [PMID: 34169956 DOI: 10.1039/d1fo01170g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Diabetic nephropathy (DN) is the most important complication in patients with diabetes. The accumulation of advanced glycation end-products (AGEs) is the main reason for the development of DN. In this study, we investigated the mechanism of buckwheat hull flavonoids to break AGEs in vitro by measuring fluorescence analysis, three-dimensional fluorescence, protein molecular weight, free amino groups, and the sulfhydryl group content. Proteomics analysis was used to determine the effect of total buckwheat hull flavonoids (TBHF) intervention on protein differential expression in the kidney of db/db mice. The results showed that buckwheat hull flavonoids were potent in breaking AGEs in vitro, and they protected mice kidneys by regulating the renal AGE-RAGE pathway. This study lays a strong experimental and theoretical foundation for the development of new lysing agents to break AGEs. The findings should make an important contribution to the field of flavonoids in improving the application of diabetic nephropathy in the diet.
Collapse
Affiliation(s)
- Tianzhu Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, Jilin, China.
| | | | | | | | | | | |
Collapse
|
30
|
Guo X, Wu Y, Zhang C, Wu L, Qin L, Liu T. Network Pharmacology Analysis of ZiShenWan for Diabetic Nephropathy and Experimental Verification of Its Anti-Inflammatory Mechanism. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1577-1594. [PMID: 33883881 PMCID: PMC8055297 DOI: 10.2147/dddt.s297683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/09/2021] [Indexed: 01/21/2023]
Abstract
Background Diabetic nephropathy (DN) is the leading cause of end-stage renal disease (ESRD). The inflammatory response plays a critical role in DN. ZiShenWan (ZSW) is a classical Chinese medicinal formula with remarkable clinical therapeutic effects on DN, but its pharmacological action mechanisms remain unclear. Aim In this study, a network pharmacology approach was applied to investigate the pharmacological mechanisms of ZSW in DN therapy. Based on the results of network analysis, the core targets and signaling pathways related to anti-inflammatory effect were verified via experiments in vivo. Methods The candidate chemical ingredients of ZSW as well as its putative targets and known therapeutic targets of DN were acquired from appropriate databases. The “herb-ingredient-target” network for ZSW in DN treatment was established. The protein–protein interaction (PPI) network of potential targets was constructed to screen the core targets. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. In addition to biochemical and pathological indicators, the core targets and signaling pathways associated with inflammation were partially validated in db/db mice at molecular level. Results A total of 56 active ingredients in ZSW and 166 DN-related targets were selected from databases. A high proportion of core targets and top signaling pathways participate in inflammation. ZSW markedly alleviated renal injuries pathologically and regulated related biomarkers. In particular, ZSW significantly inhibited the exaggerated release of inflammatory cytokines such as interleukin (IL)-1β, IL-6, tumor necrosis factor receptor (TNF)-ɑ, and monocyte chemotactic protein (MCP)-1 as well as regulating p38 mitogen-activated protein kinases (MAPK) and phosphoinositide 3-kinase (PI3K)–protein kinase B (Akt) signaling pathways in db/db mice. Conclusion This study first comprehensively investigated the active ingredients, potential targets, and molecular mechanism of ZSW as a therapy for DN. ZSW achieved renoprotective effects in DN via regulation of multiple targets and signaling pathways, especially by alleviating inflammation. Results indicate that ZSW is a promising multi-target therapeutic approach for DN treatment.
Collapse
Affiliation(s)
- Xiaoyuan Guo
- Department of Nephrology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People's Republic of China
| | - You Wu
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100078, People's Republic of China
| | - Chengfei Zhang
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100078, People's Republic of China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Lingling Qin
- Technology Department, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Tonghua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| |
Collapse
|
31
|
The PAR-1 antagonist vorapaxar ameliorates kidney injury and tubulointerstitial fibrosis. Clin Sci (Lond) 2021; 134:2873-2891. [PMID: 33078834 DOI: 10.1042/cs20200923] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/30/2022]
Abstract
Protease-activated receptor (PAR)-1 has emerged as a key profibrotic player in various organs including kidney. PAR-1 activation leads to deposition of extracellular matrix (ECM) proteins in the tubulointerstitium and induction of epithelial-mesenchymal transition (EMT) during renal fibrosis. We tested the anti-fibrotic potential of vorapaxar, a clinically approved PAR-1 antagonist for cardiovascular protection, in an experimental kidney fibrosis model of unilateral ureteral obstruction (UUO) and an AKI-to-chronic kidney disease (CKD) transition model of unilateral ischemia-reperfusion injury (UIRI), and dissected the underlying renoprotective mechanisms using rat tubular epithelial cells. PAR-1 is activated mostly in the renal tubules in both the UUO and UIRI models of renal fibrosis. Vorapaxar significantly reduced kidney injury and ameliorated morphologic changes in both models. Amelioration of kidney fibrosis was evident from down-regulation of fibronectin (Fn), collagen and α-smooth muscle actin (αSMA) in the injured kidney. Mechanistically, inhibition of PAR-1 inhibited MAPK ERK1/2 and transforming growth factor-β (TGF-β)-mediated Smad signaling, and suppressed oxidative stress, overexpression of pro-inflammatory cytokines and macrophage infiltration into the kidney. These beneficial effects were recapitulated in cultured tubular epithelial cells in which vorapaxar ameliorated thrombin- and hypoxia-induced TGF-β expression and ECM accumulation. In addition, vorapaxar mitigated capillary loss and the expression of adhesion molecules on the vascular endothelium during AKI-to-CKD transition. The PAR-1 antagonist vorapaxar protects against kidney fibrosis during UUO and UIRI. Its efficacy in human CKD in addition to CV protection warrants further investigation.
Collapse
|
32
|
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
|
33
|
Yiu WH, Li Y, Lok SWY, Chan KW, Chan LYY, Leung JCK, Lai KN, Tsu JHL, Chao J, Huang XR, Lan HY, Tang SCW. Protective role of kallistatin in renal fibrosis via modulation of Wnt/β-catenin signaling. Clin Sci (Lond) 2021; 135:429-446. [PMID: 33458750 DOI: 10.1042/cs20201161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/31/2022]
Abstract
Kallistatin is a multiple functional serine protease inhibitor that protects against vascular injury, organ damage and tumor progression. Kallistatin treatment reduces inflammation and fibrosis in the progression of chronic kidney disease (CKD), but the molecular mechanisms underlying this protective process and whether kallistatin plays an endogenous role are incompletely understood. In the present study, we observed that renal kallistatin levels were significantly lower in patients with CKD. It was also positively correlated with estimated glomerular filtration rate (eGFR) and negatively correlated with serum creatinine level. Unilateral ureteral obstruction (UUO) in animals also led to down-regulation of kallistatin protein in the kidney, and depletion of endogenous kallistatin by antibody injection resulted in aggravated renal fibrosis, which was accompanied by enhanced Wnt/β-catenin activation. Conversely, overexpression of kallistatin attenuated renal inflammation, interstitial fibroblast activation and tubular injury in UUO mice. The protective effect of kallistatin was due to the suppression of TGF-β and β-catenin signaling pathways and subsequent inhibition of epithelial-to-mesenchymal transition (EMT) in cultured tubular cells. In addition, kallistatin could inhibit TGF-β-mediated fibroblast activation via modulation of Wnt4/β-catenin signaling pathway. Therefore, endogenous kallistatin protects against renal fibrosis by modulating Wnt/β-catenin-mediated EMT and fibroblast activation. Down-regulation of kallistatin in the progression of renal fibrosis underlies its potential as a valuable clinical biomarker and therapeutic target in CKD.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ye Li
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Sarah W Y Lok
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kam Wa Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Loretta Y Y Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Joseph C K Leung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kar Neng Lai
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - James H L Tsu
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Sydney C W Tang
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
34
|
Li Y, Wang L, Xu B, Zhao L, Li L, Xu K, Tang A, Zhou S, Song L, Zhang X, Zhan H. Based on Network Pharmacology Tools to Investigate the Molecular Mechanism of Cordyceps sinensis on the Treatment of Diabetic Nephropathy. J Diabetes Res 2021; 2021:8891093. [PMID: 33628839 PMCID: PMC7884116 DOI: 10.1155/2021/8891093] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/17/2021] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the most common complications of diabetes mellitus and is a major cause of end-stage kidney disease. Cordyceps sinensis (Cordyceps, Dong Chong Xia Cao) is a widely applied ingredient for treating patients with DN in China, while the molecular mechanisms remain unclear. This study is aimed at revealing the therapeutic mechanisms of Cordyceps in DN by undertaking a network pharmacology analysis. MATERIALS AND METHODS In this study, active ingredients and associated target proteins of Cordyceps sinensis were obtained via Traditional Chinese Medicine Systems Pharmacology Database (TCMSP) and Swiss Target Prediction platform, then reconfirmed by using PubChem databases. The collection of DN-related target genes was based on DisGeNET and GeneCards databases. A DN-Cordyceps common target interaction network was carried out via the STRING database, and the results were integrated and visualized by utilizing Cytoscape software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to determine the molecular mechanisms and therapeutic effects of Cordyceps on the treatment of DN. RESULTS Seven active ingredients were screened from Cordyceps, 293 putative target genes were identified, and 85 overlapping targets matched with DN were considered potential therapeutic targets, such as TNF, MAPK1, EGFR, ACE, and CASP3. The results of GO and KEGG analyses revealed that hub targets mainly participated in the AGE-RAGE signaling pathway in diabetic complications, TNF signaling pathway, PI3K-Akt signaling pathway, and IL-17 signaling pathway. These targets were correlated with inflammatory response, apoptosis, oxidative stress, insulin resistance, and other biological processes. CONCLUSIONS Our study showed that Cordyceps is characterized as multicomponent, multitarget, and multichannel. Cordyceps may play a crucial role in the treatment of DN by targeting TNF, MAPK1, EGFR, ACE, and CASP3 signaling and involved in the inflammatory response, apoptosis, oxidative stress, and insulin resistance.
Collapse
Affiliation(s)
- Yan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Bojun Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Liangbin Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Li Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Keyang Xu
- Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang, China
| | - Anqi Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Shasha Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Lu Song
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Xiao Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| | - Huakui Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan, China
| |
Collapse
|
35
|
Exogenous pancreatic kininogenase protects against renal fibrosis in rat model of unilateral ureteral obstruction. Acta Pharmacol Sin 2020; 41:1597-1608. [PMID: 32300244 DOI: 10.1038/s41401-020-0393-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/29/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue kallikrein has protective function against various types of injury. In this study, we investigated whether exogenous pancreatic kininogenase (PK) conferred renoprotection in a rat model of unilateral ureteral obstruction (UUO) and H2O2-treated HK-2 cells in vitro. SD rats were subjected to UUO surgery, then PK (7.2 U/g per day, ip) was administered for 7 or 14 days. After the treatment, rats were euthanized; the obstructed kidneys were harvested for further examination. We found that PK administration significantly attenuated interstitial inflammation and fibrosis, and downregulated the expression of proinflammatory (MCP-1, TLR-2, and OPN) and profibrotic (TGF-β1 and CTGF) cytokines in obstructed kidney. UUO-induced oxidative stress, closely associated with excessive apoptotic cell death and autophagy via PI3K/AKT/FoxO1a signaling, which were abolished by PK administration. We further showed that PK administration increased the expression of bradykinin receptors 1 and 2 (B1R and B2R) mRNA and the production of NO and cAMP in kidney tissues. Coadministration with either B1R antagonist (des-Arg9-[Leu8]-bradykinin) or B2R antagonist (icatibant) abrogated the renoprotective effects of PK, and reduced the levels of NO and cAMP in obstructed kidney. In H2O2-treated HK-2 cells, addition of PK (6 pg/mL) significantly decreased ROS production, regulated the expression of oxidant and antioxidant enzymes, suppressed the expression of TGF-β1 and MCP-1, and inhibited cell apoptosis. Our data demonstrate that PK treatment protects against the progression of renal fibrosis in obstructed kidneys.
Collapse
|
36
|
Jiang Y, Xie F, Lv X, Wang S, Liao X, Yu Y, Dai Q, Zhang Y, Meng J, Hu G, Peng Z, Tao L. Mefunidone ameliorates diabetic kidney disease in STZ and db/db mice. FASEB J 2020; 35:e21198. [PMID: 33225469 DOI: 10.1096/fj.202001138rr] [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: 05/07/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022]
Abstract
Diabetic kidney disease (DKD) is a major cause of end stage renal diseases worldwide. Despite successive interventions for delaying the progression of DKD, current treatments cannot reverse the pathological progression. Mefunidone (MFD) is a new compound with potent antifibrotic properties, but the effect of MFD on DKD remains unknown. Therefore, we investigated the protective effects of MFD in both models of the db/db type 2 diabetes (T2D) and streptozotocin (STZ)-induced type 1 diabetes (T1D) models. Compared with the model group, MFD treatment significantly reduced pathological changes observed by PAS staining, PASM staining, and Masson staining in vivo. To further elucidate the potential mechanisms, we discovered MFD treatment notably restored podocyte function, alleviated inflammation, abated ROS generation, inhibited the TGF-β1/SAMD2/3 pathway, suppressed the phosphorylation levels of MAPKs (ERK1/2, JNK, and P38), and reduced epithelial-to-mesenchymal transition(EMT). In conclusion, these findings demonstrate the effectiveness of MFD in diabetic nephropathy and elucidate its possible mechanism.
Collapse
Affiliation(s)
- Yupeng Jiang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Feifei Xie
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Xin Lv
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Shuting Wang
- Department of Oncology, Xiangya Hospital of Central South University, Changsha, China
| | - Xiaohua Liao
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Yue Yu
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Qin Dai
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Yan Zhang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Gaoyun Hu
- Faculty of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
37
|
Loupy KM, Lee T, Zambrano CA, Elsayed AI, D'Angelo HM, Fonken LK, Frank MG, Maier SF, Lowry CA. Alzheimer's Disease: Protective Effects of Mycobacterium vaccae, a Soil-Derived Mycobacterium with Anti-Inflammatory and Anti-Tubercular Properties, on the Proteomic Profiles of Plasma and Cerebrospinal Fluid in Rats. J Alzheimers Dis 2020; 78:965-987. [PMID: 33074227 DOI: 10.3233/jad-200568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is an inflammatory neurodegenerative disease that may be associated with prior bacterial infections. Microbial "old friends" can suppress exaggerated inflammation in response to disease-causing infections or increase clearance of pathogens such as Mycobacterium tuberculosis, which causes tuberculosis (TB). One such "old friend" is Mycobacterium vaccae NCTC 11659, a soil-derived bacterium that has been proposed either as a vaccine for prevention of TB, or as immunotherapy for the treatment of TB when used alongside first line anti-TB drug treatment. OBJECTIVE The goal of this study was to use a hypothesis generating approach to explore the effects of M. vaccae on physiological changes in the plasma and cerebrospinal fluid (CSF). METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were performed in plasma and CSF of adult male rats after immunization with a heat-killed preparation of M. vaccae NCTC 11659 or borate-buffered saline vehicle. Gene enrichment analysis and analysis of protein-protein interactions were performed to integrate physiological network changes in plasma and CSF. We used RT-qPCR to assess immune and metabolic gene expression changes in the hippocampus. RESULTS In both plasma and CSF, immunization with M. vaccae increased proteins associated with immune activation and downregulated proteins corresponding to lipid (including phospholipid and cholesterol) metabolism. Immunization with M. vaccae also increased hippocampal expression of interleukin-4 (IL-4) mRNA, implicating anti-inflammatory effects in the central nervous system. CONCLUSION M. vaccae alters host immune activity and lipid metabolism. These data are consistent with the hypothesis that microbe-host interactions may protect against possible infection-induced, inflammation-related cognitive impairments.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ, USA
| |
Collapse
|
38
|
Wen S, Li S, Li L, Fan Q. circACTR2: A Novel Mechanism Regulating High Glucose-Induced Fibrosis in Renal Tubular Cells via Pyroptosis. Biol Pharm Bull 2020; 43:558-564. [PMID: 32115515 DOI: 10.1248/bpb.b19-00901] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease. Current therapies for DKD are insufficient. Therefore, there is an urgent need for identifying new therapies. An increasing number of micro RNAs (miRNAs) and long noncoding RNAs (lncRNAs) have been demonstrated to modulate the progression of diabetic kidney disease. Nevertheless, until now, there have been few reports evaluating the relevance of circular RNAs (circRNAs) in DKD. circRNAs have been reported to regulate the occurrence and development of multiple diseases. In this study, we intended to explore the circRNA expression profiles and determine the role of circRNA in DKD. We identified a series of dysregulated circRNAs in glucose-stressed HK-2 cells using circRNA microarray analysis. Among the candidate circRNAs, we found that circACTR2 was upregulated and may be involved in inflammation and pyroptosis. Knockdown of circACTR2 significantly decreased pyroptosis, interleukin (IL)-1β release and collagen IV and fibronectin production, indicating the effective regulation by circACTR2 of cell death and inflammation. Overall, our study identified a new circRNA, circACTR2, that regulates high glucose-induced pyroptosis, inflammation and fibrosis in proximal tubular cells. The present study preliminarily explores the role of circRNAs in pyroptosis of tubular cells, and provides novel insight into the pathogenesis of DKD and new therapeutic strategies.
Collapse
Affiliation(s)
- Si Wen
- Department of Nephrology, First Hospital of China Medical University
| | - Shuangliang Li
- Department of Nephrology, First Hospital of China Medical University
| | - Lulu Li
- Department of Nephrology, First Hospital of China Medical University
| | - Qiuling Fan
- Department of Nephrology, First Hospital of China Medical University
| |
Collapse
|
39
|
Huang K, Zhao X. USP9X prevents AGEs-induced upregulation of FN and TGF-β1 through activating Nrf2-ARE pathway in rat glomerular mesangial cells. Exp Cell Res 2020; 393:112100. [PMID: 32442538 DOI: 10.1016/j.yexcr.2020.112100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress is a key pathological factor for diabetic renal fibrosis by activating TGF-β/Smad pathway in glomerular mesangial cells (GMCs) to promote the synthesis of extracellular matrix such as fibronectin (FN). Nuclear factor-E2-related factor (Nrf2)- anti-oxidant response element (ARE) anti-oxidative pathway has crucial renoprotective effects, and inhibiting ubiquitin-mediated degradation of Nrf2 delays diabetic renal fibrosis development. Ubiquitin-specific protease 9X (USP9X) has close relationship with oxidative stress and TGF-β/Smad pathway, but whether it regulate diabetic renal fibrosis remains unclarified. Here, we found that advanced glycation-end products (AGEs) dose- and time-dependently reduced the protein expression and deubiquitinase activity of USP9X in GMCs. USP9X overexpression attenuated AGEs-induced upregulation of FN, TGF-β1, and Collagen Ⅳ, three fibrosis-related marker proteins, in a deubiquitinase activity-dependent manner. While USP9X depletion with siRNAs further promoted the expressions of those proteins in AGEs-treated GMCs. Under AGEs treatment conditions, USP9X overexpression markedly increased the total and nuclear levels, ARE-binding ability, and transcriptional activity of Nrf2, upregulated the protein expressions of Nrf2 downstream genes HO-1 and NQO1, and eventually reduced the excessive production of ROS. Overexpression of the deubiquitinase catalytically inactive USP9X-C1556S mutant failed to exert such effects. Silencing Nrf2 abolished the renoprotective effects of USP9X. Further study showed that upon AGEs stimulation, Nrf2 transferred into the nucleus and the interaction between USP9X and Nrf2 was weakened. AGEs also increased Nrf2 ubiquitination level, and overexpression of USP9X, instead of USP9X-C1556S, significantly reduced the ubiquitination level of Nrf2. Taken together, USP9X reduced Nrf2 ubiquitination level and promoted Nrf2-ARE pathway activation to prevent the accumulation of extracellular matrix, eventually alleviated the pathological process of diabetic renal fibrosis.
Collapse
Affiliation(s)
- Kaipeng Huang
- Phase I Clinical Trial Center, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China.
| | - Xilin Zhao
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
40
|
Du Y, Yang YT, Tang G, Jia JS, Zhu N, Yuan WJ. Butyrate alleviates diabetic kidney disease by mediating the miR-7a-5p/P311/TGF-β1 pathway. FASEB J 2020; 34:10462-10475. [PMID: 32539181 DOI: 10.1096/fj.202000431r] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/13/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022]
Abstract
It has been reported that butyrate played an protect role in diabetic kidney disease (DKD) while the mechanism was still not clear. Transforming growth factor-β1 (TGF-β1) is the initial factor which triggers the profibrotic signaling cascades. P311 is an RNA-binding protein, which could stimulate TGF-β1 translation in several cell types. In our study, we found that supplementary of butyrate alleviated fibrosis and suppressed the expression of TGF-β1 and P311 in the kidney of db/db mice as well as high glucose (HG)-induced SV40-MES-13 cells. Overexpression of P311 offset the inhibition of butyrate on TGF-β1 in SV40-MES-13 cells. To make clear the mechanism of butyrate in regulating P311, microRNAs (miRNAs) of the SV40-MES-13 cells were sequenced. We found that miR-7a-5p was significantly decreased in the HG-induced SV40-MES-13 cells and the kidney of db/db mice, while giving butyrate reversed this change. Besides, miR-7a-5p could specifically target the 3' UTR of P311's mRNA and suppressed the expression of P311 in the SV40-MES-13 cells. Giving miR-7a-5p inhibitor blocked the inhibition of butyrate on P311 and TGF-β1. Introducing the miR-7a-5p agomir into db/db mice alleviated renal fibrosis and inhibit the expression of P311 and TGF-β1. In conclusion, butyrate alleviated DKD by mediating the miR-7a-5p/P311/TGF-β1 pathway.
Collapse
Affiliation(s)
- Yi Du
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Tong Yang
- Department of Nephrology, Qingdao Municipal Hospital, Qingdao, China
| | - Gang Tang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie-Shuang Jia
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Zhu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Jie Yuan
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Muhammad F, Avalos PN, Mursalin MH, Ma JX, Callegan MC, Lee DJ. Kallistatin Attenuates Experimental Autoimmune Uveitis by Inhibiting Activation of T Cells. Front Immunol 2020; 11:975. [PMID: 32508841 PMCID: PMC7253575 DOI: 10.3389/fimmu.2020.00975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
Experimental autoimmune uveoretinitis (EAU) is a mouse model of human autoimmune uveitis. EAU spontaneously resolves and is marked by ocular autoantigen-specific regulatory immunity in the spleen. Kallikrein binding protein (KBP) or kallistatin is a serine proteinase inhibitor that inhibits angiogenesis and inflammation, but its role in autoimmune uveitis has not been explored. We report that T cells activation is inhibited and EAU is attenuated in human KBP (HKBP) mice with no significant difference in the Treg population that we previously identified both before and after recovery from EAU. Moreover, following EAU immunization HKBP mice have potent ocular autoantigen specific regulatory immunity that is functionally suppressive.
Collapse
Affiliation(s)
- Fauziyya Muhammad
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Priscilla N Avalos
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - M H Mursalin
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michelle C Callegan
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Darren J Lee
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
42
|
Tang SCW. Editorial: diabetic kidney disease: an update in recent clinical and basic research. Nephrol Dial Transplant 2020; 35:725-728. [PMID: 31327003 DOI: 10.1093/ndt/gfz130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/27/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Sydney C W Tang
- Division of Nephrology, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| |
Collapse
|
43
|
Yang Y, He X, Cheng R, Chen Q, Shan C, Chen L, Ma JX. Diabetes-induced upregulation of kallistatin levels exacerbates diabetic nephropathy via RAS activation. FASEB J 2020; 34:8428-8441. [PMID: 32352602 PMCID: PMC7302980 DOI: 10.1096/fj.201903149r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/28/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Kallistatin is an inhibitor of tissue kallikrein and also inhibits the Wnt pathway. Its role in diabetic nephropathy (DN) is uncertain. Here we reported that serum kallistatin levels were significantly increased in diabetic patients with DN compared to those in diabetic patients without DN and healthy controls, and positively correlated with urinary albumin excretion. In addition, renal kallistatin levels were significantly upregulated in mouse models of type 1 (Akita, OVE26) and type 2 diabetes (db/db). To unveil the effects of kallistatin on DN and its underlying mechanism, we crossed transgenic mice overexpressing kallistatin with OVE26 mice (KS‐tg/OVE). Kallistatin overexpression exacerbated albuminuria, renal fibrosis, inflammation, and oxidative stress in diabetes. Kallikrein activity was inhibited while the renin‐angiotensin system (RAS) upregulated in the kidney of KS‐tg/OVE mice compared to WT/OVE mice, suggesting a disturbed balance between the RAS and kallikrein‐kinin systems. As shown by immunostaining of endothelial makers, renal vascular densities were decreased accompanied by increased HIF‐1α and erythropoietin levels in the kidneys of KS‐tg/OVE mice. Taken together, high levels of kallistatin exacerbate DN at least partly by inducing RAS overactivation and hypoxia. The present study demonstrated a positive correlation between kallistatin levels and DN, suggesting a potential biomarker for prognosis of DN.
Collapse
Affiliation(s)
- Yanhui Yang
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin, China.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xuemin He
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Endocrinology and Metabolism Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rui Cheng
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Qian Chen
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Chunyan Shan
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin, China
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin, China
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
44
|
Sotokawauchi A, Nakamura N, Matsui T, Higashimoto Y, Yamagishi SI. Glyceraldehyde-Derived Pyridinium Evokes Renal Tubular Cell Damage via RAGE Interaction. Int J Mol Sci 2020; 21:ijms21072604. [PMID: 32283652 PMCID: PMC7177832 DOI: 10.3390/ijms21072604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/08/2023] Open
Abstract
Glyceraldehyde-derived advanced glycation end products (glycer-AGEs) contribute to proximal tubulopathy in diabetes. However, what glycer-AGE structure could evoke tubular cell damage remains unknown. We first examined if deleterious effects of glycer-AGEs on reactive oxygen species (ROS) generation in proximal tubular cells were blocked by DNA-aptamer that could bind to glyceraldehyde-derived pyridinium (GLAP) (GLAP-aptamer), and then investigated whether and how GLAP caused proximal tubular cell injury. GLAP-aptamer and AGE-aptamer raised against glycer-AGEs were prepared using a systemic evolution of ligands by exponential enrichment. The binding affinity of GLAP-aptamer to glycer-AGEs was measured with a bio-layer interferometry. ROS generation was evaluated using fluorescent probes. Gene expression was analyzed by reverse transcription-polymerase chain reaction (RT-PCR). GLAP-aptamer bound to glycer-AGEs with a dissociation constant of 7.7 × 10−5 M. GLAP-aptamer, glycer-AGE-aptamer, or antibodies directed against receptor for glycer-AGEs (RAGE) completely prevented glycer-AGE- or GLAP-induced increase in ROS generation, MCP-1, PAI-1, or RAGE gene expression in tubular cells. Our present results suggest that GLAP is one of the structurally distinct glycer-AGEs, which may mediate oxidative stress and inflammatory reactions in glycer-AGE-exposed tubular cells. Blockade of the interaction of GLAP-RAGE by GLAP-aptamer may be a therapeutic target for proximal tubulopathy in diabetic nephropathy.
Collapse
Affiliation(s)
- Ami Sotokawauchi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan; (A.S.); (N.N.); (T.M.)
| | - Nobutaka Nakamura
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan; (A.S.); (N.N.); (T.M.)
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan; (A.S.); (N.N.); (T.M.)
| | - Yuichiro Higashimoto
- Department of Chemistry, Kurume University School of Medicine, Kurume 830-0011, Japan;
| | - Sho-ichi Yamagishi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan
- Correspondence: ; Tel.: +81-3-3784-8693; Fax: +81-3-3784-8948
| |
Collapse
|
45
|
Mitchell T, De Miguel C, Gohar EY. Sex differences in redox homeostasis in renal disease. Redox Biol 2020; 31:101489. [PMID: 32197946 PMCID: PMC7212488 DOI: 10.1016/j.redox.2020.101489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/20/2020] [Accepted: 03/01/2020] [Indexed: 02/08/2023] Open
Abstract
Sex differences in redox signaling in the kidney present new challenges and opportunities for understanding the physiology and pathophysiology of the kidney. This review will focus on reactive oxygen species, immune-related signaling pathways and endothelin-1 as potential mediators of sex-differences in redox homeostasis in the kidney. Additionally, this review will highlight male-female differences in redox signaling in several major cardiovascular and renal disorders namely acute kidney injury, diabetic nephropathy, kidney stone disease and salt-sensitive hypertension. Furthermore, we will discuss the contribution of redox signaling in the pathogenesis of postmenopausal hypertension and preeclampsia.
Collapse
Affiliation(s)
- Tanecia Mitchell
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eman Y Gohar
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
46
|
Antioxidant Effects and Mechanisms of Medicinal Plants and Their Bioactive Compounds for the Prevention and Treatment of Type 2 Diabetes: An Updated Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1356893. [PMID: 32148647 PMCID: PMC7042557 DOI: 10.1155/2020/1356893] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/31/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus is a metabolic disorder that majorly affects the endocrine gland, and it is symbolized by hyperglycemia and glucose intolerance owing to deficient insulin secretory responses and beta cell dysfunction. This ailment affects as many as 451 million people worldwide, and it is also one of the leading causes of death. In spite of the immense advances made in the development of orthodox antidiabetic drugs, these drugs are often considered not successful for the management and treatment of T2DM due to the myriad side effects associated with them. Thus, the exploration of medicinal herbs and natural products as therapeutic sources for the treatment of T2DM is promoted because they have little or no side effects. Bioactive molecules isolated from natural sources have been proven to lower blood glucose levels via regulating one or more of the following mechanisms: improvement of beta cell function, insulin resistance, glucose (re)absorption, and glucagon-like peptide-1 homeostasis. In recent times, the mechanisms of action of different bioactive molecules with antidiabetic properties and phytochemistry are gaining a lot of attention in the area of drug discovery. This review article presents an update of the findings from clinical research into medicinal plant therapy for T2DM.
Collapse
|
47
|
Abstract
Increasing evidence suggests that renal inflammation contributes to the pathogenesis and progression of diabetic kidney disease (DKD) and that anti-inflammatory therapies might have renoprotective effects in DKD. Immune cells and resident renal cells that activate innate immunity have critical roles in triggering and sustaining inflammation in this setting. Evidence from clinical and experimental studies suggests that several innate immune pathways have potential roles in the pathogenesis and progression of DKD. Toll-like receptors detect endogenous danger-associated molecular patterns generated during diabetes and induce a sterile tubulointerstitial inflammatory response via the NF-κB signalling pathway. The NLRP3 inflammasome links sensing of metabolic stress in the diabetic kidney to activation of pro-inflammatory cascades via the induction of IL-1β and IL-18. The kallikrein-kinin system promotes inflammatory processes via the generation of bradykinins and the activation of bradykinin receptors, and activation of protease-activated receptors on kidney cells by coagulation enzymes contributes to renal inflammation and fibrosis in DKD. In addition, hyperglycaemia leads to protein glycation and activation of the complement cascade via recognition of glycated proteins by mannan-binding lectin and/or dysfunction of glycated complement regulatory proteins. Data from preclinical studies suggest that targeting these innate immune pathways could lead to novel therapies for DKD.
Collapse
|
48
|
Wang Y, Liu T, Ma F, Lu X, Mao H, Zhou W, Yang L, Li P, Zhan Y. A Network Pharmacology-Based Strategy for Unveiling the Mechanisms of Tripterygium Wilfordii Hook F against Diabetic Kidney Disease. J Diabetes Res 2020; 2020:2421631. [PMID: 33274236 PMCID: PMC7695487 DOI: 10.1155/2020/2421631] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/01/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) poses a major public-health burden globally. Tripterygium wilfordii Hook F (TwHF) is a widely employed herbal medicine in decreasing albuminuria among diabetic patients. However, a holistic network pharmacology strategy to investigate the active components and therapeutic mechanism underlying DKD is still unavailable. METHODS We collected TwHF ingredients and their targets by traditional Chinese Medicine databases (TCMSP). Then, we obtained DKD targets from GeneCards and OMIM and collected and analyzed TwHF-DKD common targets using the STRING database. Protein-protein interaction (PPI) network was established by Cytoscape and analyzed by MCODE plugin to get clusters. In addition, the cytoHubba software was used to identify hub genes. Finally, all the targets of clusters were subjected for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses via DAVID. RESULTS A total of 51 active ingredients in TwHF were identified and hit by 88 potential targets related to DKD. Compounds correspond to more targets include kaempferol, beta-sitosterol, stigmasterol, and Triptoditerpenic acid B, which appeared to be high-potential compounds. Genes with higher degree including VEGFA, PTGS2, JUN, MAPK8, and HSP90AA1 are hub genes of TwHF against DKD, which are involved in inflammation, insulin resistance, and lipid homeostasis. Kaempferol and VEGFA were represented as the uppermost active ingredient and core gene of TwHF in treating DKD, respectively. DAVID results indicated that TwHF may play a role in treating DKD through AGE-RAGE signaling pathway, IL-17 signaling pathway, TNF signaling pathway, insulin resistance, and calcium signaling pathway (P < 0.05). CONCLUSION Kaempferol and VEGFA were represented as the uppermost active ingredient and core gene of TwHF in treating DKD, respectively. The key mechanisms of TwHF against DKD might be involved in the reduction of renal inflammation by downregulating VEGFA.
Collapse
Affiliation(s)
- Yuyang Wang
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tongtong Liu
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
| | - Fang Ma
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
| | - Xiaoguang Lu
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
| | - Huimin Mao
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
| | - Weie Zhou
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Liping Yang
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
| | - Ping Li
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yongli Zhan
- Department of Nephrology, Guang'anmen Hospital of China Academy of Traditional Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
49
|
Sembach FE, Fink LN, Johansen T, Boland BB, Secher T, Thrane ST, Nielsen JC, Fosgerau K, Vrang N, Jelsing J, Pedersen TX, Østergaard MV. Impact of sex on diabetic nephropathy and the renal transcriptome in UNx db/db C57BLKS mice. Physiol Rep 2019; 7:e14333. [PMID: 31876119 PMCID: PMC6930935 DOI: 10.14814/phy2.14333] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Diabetic nephropathy (DN) is associated with albuminuria and loss of kidney function and is the leading cause of end-stage renal disease. Despite evidence of sex-associated differences in the progression of DN in human patients, male mice are predominantly being used in preclinical DN research and drug development. Here, we compared renal changes in male and female uninephrectomized (UNx) db/db C57BLKS mice using immunohistochemistry and RNA sequencing. Male and female UNx db/db mice showed similar progression of type 2 diabetes, as assessed by obesity, hyperglycemia, and HbA1c. Progression of DN was also similar between sexes as assessed by kidney and glomerular hypertrophy as well as urine albumin-to-creatinine ratio being increased in UNx db/db compared with control mice. In contrast, kidney collagen III and glomerular collagen IV were increased only in female UNx db/db as compared with respective control mice but showed a similar tendency in male UNx db/db mice. Comparison of renal cortex transcriptomes by RNA sequencing revealed 66 genes differentially expressed (p < .01) in male versus female UNx db/db mice, of which 9 genes were located on the sex chromosomes. In conclusion, male and female UNx db/db mice developed similar hallmarks of DN pathology, suggesting no or weak sex differences in the functional and structural changes during DN progression.
Collapse
Affiliation(s)
- Frederikke E. Sembach
- Gubra ApSHørsholmDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Li XQ, Chang DY, Chen M, Zhao MH. Deficiency of C3a receptor attenuates the development of diabetic nephropathy. BMJ Open Diabetes Res Care 2019; 7:e000817. [PMID: 31798904 PMCID: PMC6861086 DOI: 10.1136/bmjdrc-2019-000817] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/18/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022] Open
Abstract
Objective Diabetic nephropathy (DN) is the leading cause of chronic kidney disease and end-stage renal disease. Emerging evidence suggests that complement activation is involved in the pathogenesis of DN. The aim of this study was to investigate the pathogenic role of C3a and C3a receptor (C3aR) in DN. Research design and methods The expression of C3aR was examined in the renal specimen of patients with DN. Using a C3aR gene knockout mice (C3aR-/-), we evaluated kidney injury in diabetic mice. The mouse gene expression microarray was performed to further explore the pathogenic role of C3aR. Then the underlying mechanism was investigated in vitro with macrophage treated with C3a. Results Compared with normal controls, the renal expression of C3aR was significantly increased in patients with DN. C3aR-/- diabetic mice developed less severe diabetic renal damage compared with wild-type (WT) diabetic mice, exhibiting significantly lower level of albuminuria and milder renal pathological injury. Microarray profiling uncovered significantly suppressed inflammatory responses and T-cell adaptive immunity in C3aR-/- diabetic mice compared with WT diabetic mice, and this result was further verified by immunohistochemical staining of renal CD4+, CD8+ T cells and macrophage infiltration. In vitro study demonstrated C3a can enhance macrophage-secreted cytokines which could induce inflammatory responses and differentiation of T-cell lineage. Conclusions C3aR deficiency could attenuate diabetic renal damage through suppressing inflammatory responses and T-cell adaptive immunity, possibly by influencing macrophage-secreted cytokines. Thus, C3aR may be a promising therapeutic target for DN.
Collapse
Affiliation(s)
- Xiao-Qian Li
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Dong-Yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|