1
|
Zhong X, Zhang J. RUNX3-activated apelin signaling inhibits cell proliferation and fibrosis in diabetic nephropathy by regulation of the SIRT1/FOXO pathway. Diabetol Metab Syndr 2024; 16:167. [PMID: 39014438 PMCID: PMC11253400 DOI: 10.1186/s13098-024-01393-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Diabetic nephropathy is a major secondary cause of end-stage renal disease. Apelin plays an important role in the development of DN. Understanding the exact mechanism of Apelin can help expand the means of treating DN. METHODS Male C57BL/6 mice was used and STZ treatment was implemented for DN model establishment. Lentivirus systems including Lv-sh-RUNX3 and Lv-Apelin were obtained to knockdown RUNX3 and overexpress Apelin, respectively. A total of 36 mice were divided into 6 groups (n = 6 in each group): control, DN, DN + LV-Vector, DN + Lv-Apelin, DN + LV-Apelin + LV-sh-NC and DN + Lv-Apelin + Lv-sh-RUNX3 group. In vitro studies were performed using mesangial cells. Cell viability and proliferation was assessed through CCK8 and EDU analysis. Hematoxylin and eosin staining as well as Masson staining was implemented for histological evaluation. RT-qPCR was conducted for measuring relative mRNA levels, and protein expression was detected by western blotting. The interaction between SIRT1 and FOXO were verified by co-immunoprecipitations, and relations between RUNX3 and Apelin were demonstrated by dual luciferase report and chromatin immunoprecipitation. RESULTS The DN group exhibited significantly lower Apelin expression compared to control (p < 0.05). Apelin overexpression markedly improved blood glucose, renal function indicators, ameliorated renal fibrosis and reduced fibrotic factor expression (p < 0.05) in the DN group, accompanied by elevated sirt1 levels and diminished acetylated FOXO1/FOXO3a (p < 0.05). However, RUNX3 knockdown combined with Apelin overexpression abrogated these beneficial effects, leading to impaired renal function, exacerbated fibrosis, increased fibrotic factor expression and acetylated FOXO1/FOXO3a versus Apelin overexpression alone (p < 0.05). In mesangial cells under high glucose, Apelin overexpression significantly inhibited cell proliferation and fibrotic factor production (p < 0.05). Conversely, RUNX3 interference enhanced cell proliferation and the secretion of fibrotic factors. (p < 0.05). Remarkably, combining Apelin overexpression with RUNX3 interference reversed the proliferation and fibrosis induced by RUNX3 interference (p < 0.05). Mechanistic studies revealed RUNX3 binds to the Apelin promoter, with the 467-489 bp site1 as the primary binding region, and SIRT1 physically interacts with FOXO1 and FOXO3a in mesangial cells. CONCLUSION RUNX3 activated Apelin and regulated the SIRT1/FOXO signaling pathway, resulting in the suppressed cell proliferation and fibrosis in diabetic nephropathy. Apelin is a promising endogenous therapeutic target for anti-renal injury and anti-fibrosis in diabetic nephropathy. RUNX3 may serve as an endogenous intervention target for diseases related to Apelin deficiency.
Collapse
Affiliation(s)
- Xin Zhong
- Department of Nephrology, The Second Clinical Medical College), Zhujiang Hospital of Southern Medical University, No. 253, Middle Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, People's Republic of China
- Department of Nephrology, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong Province, People's Republic of China
| | - Jun Zhang
- Department of Nephrology, The Second Clinical Medical College), Zhujiang Hospital of Southern Medical University, No. 253, Middle Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, People's Republic of China.
| |
Collapse
|
2
|
Yang N, Zhang Y, Ren P, Zhao L, Zheng D, Fu L, Jin J. LncRNA AA465934 Improves Podocyte Injury by Promoting Tristetraprolin-Mediated HMGB1 DownRegulation in Diabetic Nephropathy. Mol Cell Biol 2024; 44:87-102. [PMID: 38520226 PMCID: PMC10986766 DOI: 10.1080/10985549.2024.2325527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 03/25/2024] Open
Abstract
Although LncRNA AA465934 expression is reduced in high glucose (HG)-treated podocytes, its role in HG-mediated podocyte injury and diabetic nephropathy (DN) remains unknown. Herein, we investigated the role of AA465934 in HG-mediated podocyte injury and DN using a spontaneous type II diabetic nephropathy (T2DN) model. The model was created by injecting AA465934 overexpressed adeno-associated virus (AAV) or control into mice. The levels of renal function, proteinuria, renal structural lesions, and podocyte apoptosis were then examined. Furthermore, AA465934 and autophagy levels, as well as tristetraprolin (TTP) and high mobility group box 1 (HMGB1) expression changes were detected. We also observed podocyte injury and the binding ability of TTP to E3 ligase proviral insertion in murine lymphomas 2 (PIM2), AA465934, or HMGB1. According to the results, AA465934 improved DN progression and podocyte damage in T2DN mice. In addition, AA465934 bound to TTP and inhibited its degradation by blocking TTP-PIM2 binding. Notably, TTP knock-down blocked the ameliorating effects of AA465934 and TTP bound HMGB1 mRNA, reducing its expression. Overexpression of HMGB1 inhibited the ability of AA465934 and TTP to improve podocyte injury. Furthermore, AA465934 bound TTP, inhibiting TTP-PIM2 binding, thereby suppressing TTP degradation, downregulating HMGB1, and reversing autophagy downregulation, ultimately alleviating HG-mediated podocyte injury and DN. Based on these findings, we deduced that the AA465934/TTP/HMGB1/autophagy axis could be a therapeutic avenue for managing podocyte injury and DN.
Collapse
Affiliation(s)
- Nan Yang
- Postgraduate Training Base of Jinzhou Medical University (Zhejiang Provincial People’s Hospital), Jinzhou, Liaoning, China
| | - Yue Zhang
- The Medical College of Qingdao University, Qingdao, China
| | - Peiyao Ren
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Li Zhao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Danna Zheng
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lanjun Fu
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Juan Jin
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Xing C, Huo L, Tang H, Lu Y, Liu G, Chen F, Hou Z. The predictive value of miR-377 and phospholipase A2 in the early diagnosis of diabetic kidney disease and their relationship with inflammatory factors. Immunobiology 2024; 229:152792. [PMID: 38401467 DOI: 10.1016/j.imbio.2024.152792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/02/2024] [Accepted: 02/11/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVE The value of novel biomarkers for DKD has received increasing attention, and there is an urgent need for novel biomarkers with sensitivity, specificity and ability to detect kidney damage.miR-377 regulates many basic biological processes, plays a key role in tumor cell proliferation, migration and inflammation, and can also increase the expression of matrix proteins and fibronectin, leading to renal tubulointerstitial inflammation and renal fibrosis. Lipoprotein-associated phospholipase A2, as an inflammatory marker, is involved in the pathological process of microalbuminuria production and renal function decline, and is a predictive factor of microalbuminuria production and renal function decline, and can be used as an indicator to evaluate the progression of DKD.The aim of this study was to investigate the effects of miR-377 and phospholipase A2 on the development of diabetic kidney disease through regulation of inflammatory factors and the mechanism of action. METHODS 80 diabetic patients were divided into two groups according to urinary albumin-to-creatinine ratio (UACR): diabetic normal proteinuria group (n = 42) and diabetic proteinuria group (n = 38). Forty-three healthy people were selected as the normal control group. The serum levels of TGF-β, IL-6, and IL-18 were measured by ELISA, miR-377 was detected by qPCR, and the serum levels of phospholipase A2 were detected by electrochemiluminescence. Analyze the correlation of study group indicators, ROC curve was used to evaluate the diagnostic efficacy of miR-377 and phospholipase A2 in diabetic kidney disease. RESULTS The average levels of serum TGF-β, IL-6, IL-18, miR-377 and phospholipase A2 in diabetic proteinuria group were significantly higher than those in normal control group and diabetic proteinuria normal group(P < 0.05). miR-377, phospholipase A2 were significantly correlated with inflammatory factors such as glomerular filtration rate and TGF-β. miR-377 and phospholipase A2 are independent predictors of diabetic kidney disease. The area under the curve of miR-377 and phospholipase A2 in the normal diabetic proteinuria group and the diabetic proteinuria group were 0.731 and 0.744, respectively. CONCLUSION miR-377 and phospholipase A2 have good diagnostic efficiency for the early diagnosis of diabetic kidney disease. They can be used as early biomarkers.miR-377 and phospholipase A2 were positively correlated with inflammatory factors and involved in the occurrence and development of diabetic kidney disease.
Collapse
Affiliation(s)
- Chenhao Xing
- Hebei North University, Zhang Jiakou 075000, Hebei province, China
| | - Lijing Huo
- Clinical laboratory of Hebei General Hospital, Shijiazhuang 050051, Hebei province, China
| | - Hongyue Tang
- Hebei North University, Zhang Jiakou 075000, Hebei province, China
| | - Yamin Lu
- Department of Nuclear Medicine of Hebei General Hospital, Shijiazhuang 050051, Hebei province, China.
| | - Guangxia Liu
- Department of Nuclear Medicine of Hebei General Hospital, Shijiazhuang 050051, Hebei province, China
| | - Fang Chen
- Department of Nuclear Medicine of Hebei General Hospital, Shijiazhuang 050051, Hebei province, China
| | - Zhan Hou
- Department of Nuclear Medicine of Hebei General Hospital, Shijiazhuang 050051, Hebei province, China
| |
Collapse
|
4
|
Peng R, Zuo S, Li X, Huang Y, Chen S, Zou X, Long H, Chen M, Yang Y, Yuan H, Zhao Q, Guo B, Liu L. Investigating HMGB1 as a potential serum biomarker for early diabetic nephropathy monitoring by quantitative proteomics. iScience 2024; 27:108834. [PMID: 38303703 PMCID: PMC10830865 DOI: 10.1016/j.isci.2024.108834] [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: 09/01/2023] [Revised: 12/01/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Current diagnostic methods for diabetic nephropathy (DN) lack precision, especially in early stages and monitoring progression. This study aims to find potential biomarkers for DN progression and evaluate their accuracy. Using serum samples from healthy controls (NC), diabetic patients (DM), early-medium stage DN (DN-EM), and late-stage DN (DN-L), researchers employed quantitative proteomics and Mfuzz clustering analysis revealed 15 proteins showing increased expression during DN progression, hinting at their biomarker potential. Combining Mfuzz clustering with weighted gene co-expression network analysis (WGCNA) highlighted five candidates (HMGB1, CD44, FBLN1, PTPRG, and ADAMTSL4). HMGB1 emerged as a promising biomarker, closely correlated with renal function changes. Experimental validation supported HMGB1's upregulation under high glucose conditions, reinforcing its potential as an early detection biomarker for DN. This research advances DN understanding and identifies five potential biomarkers, notably HMGB1, as a promising early monitoring target. These findings set the stage for future clinical diagnostic applications in DN.
Collapse
Affiliation(s)
- Rui Peng
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Siyang Zuo
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Xia Li
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- Center for Clinical Medical Research, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yun Huang
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Siyu Chen
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Xue Zou
- Center for Clinical Medical Research, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Hehua Long
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Min Chen
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yuan Yang
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Huixiong Yuan
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qingqing Zhao
- Center for Clinical Medical Research, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, China
- Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang 550025, China
| | - Lirong Liu
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
- Guizhou Precision Medicine Institute, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
5
|
Hadpech S, Thongboonkerd V. Epithelial-mesenchymal plasticity in kidney fibrosis. Genesis 2024; 62:e23529. [PMID: 37345818 DOI: 10.1002/dvg.23529] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is an important biological process contributing to kidney fibrosis and chronic kidney disease. This process is characterized by decreased epithelial phenotypes/markers and increased mesenchymal phenotypes/markers. Tubular epithelial cells (TECs) are commonly susceptible to EMT by various stimuli, for example, transforming growth factor-β (TGF-β), cellular communication network factor 2, angiotensin-II, fibroblast growth factor-2, oncostatin M, matrix metalloproteinase-2, tissue plasminogen activator (t-PA), plasmin, interleukin-1β, and reactive oxygen species. Similarly, glomerular podocytes can undergo EMT via these stimuli and by high glucose condition in diabetic kidney disease. EMT of TECs and podocytes leads to tubulointerstitial fibrosis and glomerulosclerosis, respectively. Signaling pathways involved in EMT-mediated kidney fibrosis are diverse and complex. TGF-β1/Smad and Wnt/β-catenin pathways are the major venues triggering EMT in TECs and podocytes. These two pathways thus serve as the major therapeutic targets against EMT-mediated kidney fibrosis. To date, a number of EMT inhibitors have been identified and characterized. As expected, the majority of these EMT inhibitors affect TGF-β1/Smad and Wnt/β-catenin pathways. In addition to kidney fibrosis, these EMT-targeted antifibrotic inhibitors are expected to be effective for treatment against fibrosis in other organs/tissues.
Collapse
Affiliation(s)
- Sudarat Hadpech
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
6
|
Ren H, Huang M, Ou L, Deng X, Wu X, Gong Q, Liu B. Autophagy inhibitor 3-methyladenine attenuates renal injury in streptozotocin-induced diabetic mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:793-800. [PMID: 38800022 PMCID: PMC11127078 DOI: 10.22038/ijbms.2024.71378.15518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 11/13/2023] [Indexed: 05/29/2024]
Abstract
Objectives To investigate whether 3-methyladenine (3-MA) can protect the kidney of streptozotocin (STZ) - induced diabetes mice, and explore its possible mechanism. Materials and Methods STZ was used to induce diabetes in C57BL/6J mice. The mice were divided into normal control group (NC), diabetes group (DM), and diabetes+3-MA intervention group (DM+3-MA). Blood glucose, water consumption, and body weight were recorded weekly. At the end of the 6th week of drug treatment, 24-hour urine was collected. Blood and kidneys were collected for PAS staining to evaluate the degree of renal injury. Sirius red staining was used to assess collagen deposition. Blood urea nitrogen (BUN), serum creatinine, and 24-hour urine albumin were used to evaluate renal function. Western blot was used to detect fibrosis-related protein, inflammatory mediators, high mobility group box 1 (HMGB1)/NF-κB signal pathway molecule, vascular endothelial growth factor (VEGF), and podocin, and immunohistochemistry (IHC) was used to detect the expression and localization of autophagy-related protein and fibronectin. Results Compared with the kidney of normal control mice, the kidney of diabetes control mice was more pale and hypertrophic. Hyperglycemia induces renal autophagy and activates the HMGB1/NF-κB signal pathway, leading to the increase of inflammatory mediators, extracellular matrix (ECM) deposition, and proteinuria in the kidney. In diabetic mice treated with 3-MA, blood glucose decreased, autophagy and HMGB1/NF-κB signaling pathways in the kidneys were inhibited, and proteinuria, renal hypertrophy, inflammation, and fibrosis were improved. Conclusion 3-MA can attenuate renal injury in STZ-induced diabetic mice through inhibition of autophagy and HMGB1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Haiwen Ren
- Department of Clinical Laboratory, Bishan Hospital of Chongqing Medical University, Chongqing 402760, China
- Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Mengxin Huang
- Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Liwen Ou
- Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Xuan Deng
- Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Xin Wu
- Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Quan Gong
- Department of Immunology, Medical School of Yangtze University, Jingzhou 434023, China
- Clinical Molecular Immunology Center, Medical School of Yangtze University, Jingzhou 434023, China
| | - Benju Liu
- Department of Human Anatomy, Medical School of Yangtze University, Jingzhou 434023, China
| |
Collapse
|
7
|
Xu C, Chen Y, Liu Z, Fu X. Hedysarum polybotrys polysaccharide attenuates renal inflammatory infiltration and fibrosis in diabetic mice by inhibiting the HMGB1/RAGE/TLR4 pathway. Exp Ther Med 2023; 26:493. [PMID: 37771649 PMCID: PMC10523352 DOI: 10.3892/etm.2023.12192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/31/2023] [Indexed: 09/30/2023] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of kidney failure. Previous studies demonstrated the therapeutic potential of Astragalus polysaccharide in treating diabetic nephropathy. Astragalus and Hongqi both come from the leguminous plant Astragalus, but their species and genera are different, belonging to the same family and different genera of traditional Chinese medicinal plants. However, the effects of Hedysarum polybotrys polysaccharide (HPS), a polysaccharide compound from Hongqi, on DKD, including its components and efficacy, have remained elusive. The present study utilized db/db mice as a DKD animal model administered with low (30 mg/kg) and high doses (60 mg/kg) of HPS, in addition to glyburide (7.2 mg/kg). Blood and urine samples were collected from mice and blood glucose, serum creatinine, urinary albumin excretion and urinary β2-microglobulin were measured. In addition, apoptosis and histological changes in kidney tissue were observed using TUNEL and HE staining, respectively, and the secretion and expression of inflammatory factors in kidney tissue were detected using EILSA and reverse transcription-quantitative PCR. Furthermore, we the expression of fibrosis-related proteins and NF-κB signaling pathway proteins was determined using western blot analysis. HPS was found to reduce the blood glucose concentration, serum creatinine levels, urinary albumin excretion rates and urinary β2-microglobulin in a dose-dependent manner. In addition, HPS treatment mitigated apoptosis and pathological damage in the kidney tissues of DKD mice. The expression levels of fibrosis-related proteins fibronectin, α-smooth muscle actin and TGF-β1 were observed to be decreased in kidney tissues of DKD mice following HPS treatment. The secretion levels of inflammatory factors (IL-6, TNF-α and IL-1β) were also reduced in kidney tissues, with high-dose HPS treatment found to be more effective, similar to the effects mediated by the glyburide. Further mechanistic analysis revealed that the therapeutic effects of HPS on DKD mice may be mediated by inhibiting the high mobility group box 1/receptor for advanced glycation end-products/toll-like receptor 4 pathway. In conclusion, the present findings could provide insight for the treatment of DKD.
Collapse
Affiliation(s)
- Changqing Xu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Yanxu Chen
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Zongmei Liu
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 710077, P.R. China
| | - Xiaoyan Fu
- Hyperbaric Oxygen Chamber, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 710077, P.R. China
| |
Collapse
|
8
|
Liu T, Li Q, Jin Q, Yang L, Mao H, Qu P, Guo J, Zhang B, Ma F, Wang Y, Peng L, Li P, Zhan Y. Targeting HMGB1: A Potential Therapeutic Strategy for Chronic Kidney Disease. Int J Biol Sci 2023; 19:5020-5035. [PMID: 37781525 PMCID: PMC10539693 DOI: 10.7150/ijbs.87964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023] Open
Abstract
High-mobility group protein box 1 (HMGB1) is a member of a highly conserved high-mobility group protein present in all cell types. HMGB1 plays multiple roles both inside and outside the cell, depending on its subcellular localization, context, and post-translational modifications. HMGB1 is also associated with the progression of various diseases. Particularly, HMGB1 plays a critical role in CKD progression and prognosis. HMGB1 participates in multiple key events in CKD progression by activating downstream signals, including renal inflammation, the onset of persistent fibrosis, renal aging, AKI-to-CKD transition, and important cardiovascular complications. More importantly, HMGB1 plays a distinct role in the chronic pathophysiology of kidney disease, which differs from that in acute lesions. This review describes the regulatory role of HMGB1 in renal homeostasis and summarizes how HMGB1 affects CKD progression and prognosis. Finally, some promising therapeutic strategies for the targeted inhibition of HMGB1 in improving CKD are summarized. Although the application of HMGB1 as a therapeutic target in CKD faces some challenges, a more in-depth understanding of the intracellular and extracellular regulatory mechanisms of HMGB1 that underly the occurrence and progression of CKD might render HMGB1 an attractive therapeutic target for CKD.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Qu
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Zhang
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Fu Y, Xiang Y, Wang Y, Liu Z, Yang D, Zha J, Tang C, Cai J, Chen G, Dong Z. The STAT1/HMGB1/NF-κB pathway in chronic inflammation and kidney injury after cisplatin exposure. Theranostics 2023; 13:2757-2773. [PMID: 37284446 PMCID: PMC10240827 DOI: 10.7150/thno.81406] [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: 11/30/2022] [Accepted: 04/26/2023] [Indexed: 06/08/2023] Open
Abstract
Rationale: Cisplatin, a potent chemotherapeutic drug, induces side effects in normal tissues including the kidney. To reduce the side effects, repeated low-dose cisplatin (RLDC) is commonly used in clinical setting. While RLDC reduces acute nephrotoxicity to certain extents, a significant portion of patients later develop chronic kidney problems, underscoring the need for novel therapeutics to alleviate the long-term sequelae of RLDC therapy. Methods: In vivo, the role of HMGB1 was examined by testing HMGB1 neutralizing antibodies in RLDC mice. In vitro, the effects of HMGB1 knockdown on RLDC-induced nuclear factor-κB (NF-κB) activation and fibrotic phenotype changes were tested in proximal tubular cells. To study signal transducer and activator of transcription 1 (STAT1), siRNA knockdown and its pharmacological inhibitor Fludarabine were used. We also searched the Gene Expression Omnibus (GEO) database for transcriptional expression profiles and evaluated kidney biopsy samples from CKD patients to verify the STAT1/HMGB1/NF-κB signaling axis. Results: We found that RLDC induced kidney tubule damage, interstitial inflammation, and fibrosis in mice, accompanied by up-regulation of HMGB1. Blockage of HMGB1with neutralizing antibodies and Glycyrrhizin suppressed NF-κB activation and associated production of pro-inflammatory cytokines, reduced tubular injury and renal fibrosis, and improved renal function after RLDC treatment. Consistently, knockdown of HMGB1 decreased NF-κB activation and prevented the fibrotic phenotype in RLDC-treated renal tubular cells. At the upstream, knockdown of STAT1 suppressed HMGB1 transcription and cytoplasmic accumulation in renal tubular cells, suggesting a critical role of STAT1 in HMGB1 activation. Upregulation of STAT1/HMGB1/NF-κB along with inflammatory cytokines was also verified in kidney tissues of CKD patients. Conclusion: These results unravel the STAT1/HMGB1/NF-κB pathway that contributes to persistent inflammation and chronic kidney problems after cisplatin nephrotoxicity, suggesting new therapeutic targets for kidney protection in cancer patients receiving cisplatin chemotherapy.
Collapse
Affiliation(s)
- Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Ying Wang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhiwen Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Danyi Yang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Jie Zha
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Guochun Chen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| |
Collapse
|
10
|
Huang Y, Wang A, Jin S, Liu F, Xu F. Activation of the NLRP3 inflammasome by HMGB1 through inhibition of the Nrf2/HO-1 pathway promotes bleomycin-induced pulmonary fibrosis after acute lung injury in rats. Allergol Immunopathol (Madr) 2023; 51:56-67. [PMID: 37169561 DOI: 10.15586/aei.v51i3.668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/23/2022] [Indexed: 05/13/2023]
Abstract
OBJECTIVE Acute lung injury (ALI) is a common complication of critical diseases with high morbidity and mortality. This study explored the regulatory role and mechanism of high mobility histone box 1 protein (HMGB1) on pulmonary fibrosis (PF) after ALI in rats through nucleotide oligomerization domain-like receptor protein-3 (NLRP3) inflammasome. METHODS PF rat models after ALI were established by induction of bleomycin. Degree of fibrosis was assessed by Masson staining and Ashcroft scoring. Hydroxyproline (Hyp) contents in lung tissues and rat lung tissue morphology were detected by enzyme-linked-immunosorbent serologic assay (ELISA) and hematoxylin and eosin staining. The levels of NLRP3, major proteins of NLRP3 inflammasome (NLRP3/ASC/caspase-1), and downstream inflammatory cytokines interleukin (IL)-1 and IL-18 were determined using immunohistochemistry, Western blotting analysis, and ELISA. The nuclear/cytoplasmic nuclear factor erythroid 2-related factor 2 (Nrf2) levels and HO-1 levels were measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blotting analysis. Rats was injected with lentivirus carrying short hairpin (sh)-HMGB1 and zinc protoporphyria (ZNPP) (HO-1 inhibitor) to assess the effects of HMGB1 and HO-1 on PF and NLRP3 inflammasome activation. RESULTS Bleomycin induced PF after ALI in rats, manifested as patchy fibrosis, atelectasis, and excessive expansion, and increased Aschcroft score and Hyp content. Bleomycin treatment enhanced levels of NLRP3, ASC, caspase-1, IL-1, and IL-18 in rat lung tissues, which promoted activation of NLRP3 inflammasome. HMGB1 was up-regulated in bleomycin-induced rats. HMGB1 knockdown partially reversed NLRP3 inflammasome activation and PF progression. HMGB1 knockdown promoted Nrf2 nuclear translocation and up-regulated HO-1. Suppression of HO-1 partially reversed inhibition of HMGB1 knockdown on NLRP3 inflammasome activation and PF. CONCLUSION HMGB1 can activate NLRP3 inflammasomes and promote PF by inhibiting the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Ying Huang
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, Hubei Province, China
| | - Aili Wang
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, Hubei Province, China
| | - Sheng Jin
- Nephrology Department of Integrated Traditional Chinese and Western Medicine, Hubei No. 3 People's Hospital of Jianghan University, Wuhan 430033, Hubei Province, China
| | - Fang Liu
- Department of Respiratory Medicine, Hubei No. 3 People's Hospital of Jianghan University, Wuhan 430033, Hubei Province, China;
| | - Fang Xu
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, Hubei Province, China;
| |
Collapse
|
11
|
Gaboriaud C, Lorvellec M, Rossi V, Dumestre-Pérard C, Thielens NM. Complement System and Alarmin HMGB1 Crosstalk: For Better or Worse. Front Immunol 2022; 13:869720. [PMID: 35572583 PMCID: PMC9095977 DOI: 10.3389/fimmu.2022.869720] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/04/2022] [Indexed: 12/21/2022] Open
Abstract
Our immune system responds to infectious (PAMPs) and tissue damage (DAMPs) signals. The complement system and alarmin High-Mobility Group Box 1 (HMGB1) are two powerful soluble actors of human host defense and immune surveillance. These systems involve molecular cascades and amplification loops for their signaling or activation. Initially activated as alarm raising systems, their function can be finally switched towards inflammation resolution, where they sustain immune maturation and orchestrate repair mechanisms, opening the way back to homeostasis. However, when getting out of control, these defense systems can become deleterious and trigger serious cellular and tissue damage. Therefore, they can be considered as double-edged swords. The close interaction between the complement and HMGB1 pathways is described here, as well as their traditional and non-canonical roles, their functioning at different locations and their independent and collective impact in different systems both in health and disease. Starting from these systems and interplay at the molecular level (when elucidated), we then provide disease examples to better illustrate the signs and consequences of their roles and interaction, highlighting their importance and possible vicious circles in alarm raising and inflammation, both individually or in combination. Although this integrated view may open new therapeutic strategies, future challenges have to be faced because of the remaining unknowns regarding the molecular mechanisms underlying the fragile molecular balance which can drift towards disease or return to homeostasis, as briefly discussed at the end.
Collapse
Affiliation(s)
| | | | | | - Chantal Dumestre-Pérard
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
- Laboratoire d’Immunologie, Pôle de Biologie, CHU Grenoble Alpes, Grenoble, France
| | | |
Collapse
|
12
|
Opazo-Ríos L, Tejera-Muñoz A, Soto Catalan M, Marchant V, Lavoz C, Mas Fontao S, Moreno JA, Fierro Fernandez M, Ramos R, Suarez-Alvarez B, López-Larrea C, Ruiz-Ortega M, Egido J, Rodrigues-Díez RR. Kidney microRNA Expression Pattern in Type 2 Diabetic Nephropathy in BTBR Ob/Ob Mice. Front Pharmacol 2022; 13:778776. [PMID: 35370692 PMCID: PMC8966705 DOI: 10.3389/fphar.2022.778776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/10/2022] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy (DN) is the main leading cause of chronic kidney disease worldwide. Although remarkable therapeutic advances have been made during the last few years, there still exists a high residual risk of disease progression to end-stage renal failure. To further understand the pathogenesis of tissue injury in this disease, by means of the Next-Generation Sequencing, we have studied the microRNA (miRNA) differential expression pattern in kidneys of Black and Tan Brachyury (BTBR) ob/ob (leptin deficiency mutation) mouse. This experimental model of type 2 diabetes and obesity recapitulates the key histopathological features described in advanced human DN and therefore can provide potential useful translational information. The miRNA-seq analysis, performed in the renal cortex of 22-week-old BTBR ob/ob mice, pointed out a set of 99 miRNAs significantly increased compared to non-diabetic, non-obese control mice of the same age, whereas no miRNAs were significantly decreased. Among them, miR-802, miR-34a, miR-132, miR-101a, and mir-379 were the most upregulated ones in diabetic kidneys. The in silico prediction of potential targets for the 99 miRNAs highlighted inflammatory and immune processes, as the most relevant pathways, emphasizing the importance of inflammation in the pathogenesis of kidney damage associated to diabetes. Other identified top canonical pathways were adipogenesis (related with ectopic fatty accumulation), necroptosis (an inflammatory and regulated form of cell death), and epithelial-to-mesenchymal transition, the latter supporting the importance of tubular cell phenotype changes in the pathogenesis of DN. These findings could facilitate a better understanding of this complex disease and potentially open new avenues for the design of novel therapeutic approaches to DN.
Collapse
Affiliation(s)
- Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Universidad Autónoma de Madrid, Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad de Las Américas, Concepción, Chile
| | - Antonio Tejera-Muñoz
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Manuel Soto Catalan
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Vanessa Marchant
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Carolina Lavoz
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Sebastián Mas Fontao
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Antonio Moreno
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Maimónides Biomedical Research Institute of Cordoba (IMIBIC), UGC Nephrology, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Marta Fierro Fernandez
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Viral Vectors Service, Madrid, Spain
| | - Ricardo Ramos
- Unidad de Genómica Fundación Parque Científico de Madrid, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology Laboratory, Health Research Institute of Asturias (ISPA), Oviedo, Spain
| | - Carlos López-Larrea
- Translational Immunology Laboratory, Health Research Institute of Asturias (ISPA), Oviedo, Spain
- Department of Immunology, Hospital Universitario Central De Asturias, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Raúl R. Rodrigues-Díez
- Molecular and Cellular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Translational Immunology Laboratory, Health Research Institute of Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
13
|
Xu W, Zhang H, Zhang Q, Xu J. β-Amyrin ameliorates diabetic nephropathy in mice and regulates the miR-181b-5p/HMGB2 axis in high glucose-stimulated HK-2 cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:637-649. [PMID: 34894065 DOI: 10.1002/tox.23431] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/22/2021] [Accepted: 11/27/2021] [Indexed: 06/14/2023]
Abstract
Diabetic nephropathy (DN) is a diabetic complication that can cause renal failure. β-amyrin has been identified to possess anti-diabetic property. This study was designed to evaluate the potential role of β-amyrin in DN and its underlying mechanism. Streptozotocin-induced diabetic mice were used as the in vivo model, and high glucose (HG)-stimulated human proximal tubular HK-2 cells were utilized as the in vitro model. Renal histological changes in mice were assessed by hematoxylin-eosin and periodic acid-Schiff staining. HK-2 cell viability and apoptosis were detected by Cell Counting Kit-8 assay and flow cytometry analysis, respectively. β-amyrin was found to ameliorate kidney injury in DN mice and suppressed inflammatory response as well as apoptosis of HG-stimulated HK-2 cells. miR-181-5p expression in murine renal tissues and HK-2 cells was detected by in situ hybridization (ISH) and fluorescence in situ hybridization (FISH). MiR-181b-5p, a previously identified target for diabetic kidney disease, was downregulated in renal tissues and HG stimulated HK-2 cells, and β-amyrin induced the upregulation of miR-181b-5p. Binding relationship between miR-181b-5p and high mobility group box 2 (HMGB2) was confirmed by luciferase reporter assay. MiR-181b-5p bound to 3' untranslated region of HMGB2 to suppress its expression. As shown by immunohistochemical staining and immunofluorescence staining, HMGB2 was upregulated in the in vivo and in vitro models of DN, and β-amyrin induced the downregulation of HMGB2. Moreover, HMGB2 overexpression neutralized the suppressive effects of miR-181b-5p elevation on the inflammatory response and apoptosis of HG-treated HK-2 cells. Overall, β-amyrin ameliorates DN in mice and suppresses inflammatory response and apoptosis of HG-stimulated HK-2 cells via the miR-181b-5p/HMGB2 axis.
Collapse
Affiliation(s)
- Wenhua Xu
- Preventive Treatment Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Hongwu Zhang
- Department of Endocrinology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Qinfeng Zhang
- Department of Endocrinology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Jialan Xu
- Preventive Treatment Center, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
14
|
Zhong H, Gui X, Hou L, Lv R, Jin Y. From Inflammation to Fibrosis: Novel Insights into the Roles of High Mobility Group Protein Box 1 in Schistosome-Induced Liver Damage. Pathogens 2022; 11:pathogens11030289. [PMID: 35335612 PMCID: PMC8951358 DOI: 10.3390/pathogens11030289] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/20/2022] Open
Abstract
Schistosomiasis is a chronic helminthic disease of both humans and animals and the second most prevalent parasitic disease after malaria. Through a complex migration process, schistosome eggs trapped in the liver can lead to the formation of granulomas and subsequent schistosome-induced liver damage, which results in high mortality and morbidity. Although praziquantel can eliminate mature worms and prevent egg deposition, effective drugs to reverse schistosome-induced liver damage are scarce. High mobility group box 1 (HMGB1) is a multifunctional cytokine contributing to liver injury, inflammation, and immune responses in schistosomiasis by binding to cell-surface Toll-like receptors and receptors for advanced glycation end products. HMGB1 is increased in the serum of patients with schistosomiasis and enables hepatic stellate cells to adopt a proliferative myofibroblast-like phenotype, which is crucial to schistosome-induced granuloma formation. Inhibition of HMGB1 was found to generate protective responses against fibrotic diseases in animal models. Clinically, HMGB1 presents a potential target for treatment of the chronic sequelae of schistosomiasis. Here, the pivotal role of HMGB1 in granuloma formation and schistosome-induced liver damage, as well the potential of HMGB1 as a therapeutic target, are discussed.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (X.G.); (L.H.); (R.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xiang Gui
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (X.G.); (L.H.); (R.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Ling Hou
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (X.G.); (L.H.); (R.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030031, China
| | - Rongxue Lv
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (X.G.); (L.H.); (R.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (X.G.); (L.H.); (R.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Correspondence: ; Tel./Fax: +86-021-34293150
| |
Collapse
|
15
|
Pan Y, Liu L, Yang H, Chen W, Chen Z, Xu J. Sacubitril/Valsartan Improves Progression of Early Diabetic Nephropathy in Rats Through Inhibition of NLRP3 Inflammasome Pathway. Diabetes Metab Syndr Obes 2022; 15:2479-2488. [PMID: 35992034 PMCID: PMC9386175 DOI: 10.2147/dmso.s366518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Diabetic nephropathy (DN), a global disease, is the leading cause of end-stage renal disease. There is a lack of specific treatment for this disease, and early intervention in disease progression is essential. In this paper, we used a rat model of early diabetic nephropathy to explore the therapeutic mechanism of sacubitril/valsartan in rats with early diabetic nephropathy. MATERIALS AND METHODS Rats were grouped into 1 normal group; 2. Model group (DN group): STZ (45 mg/kg/d) induced early diabetic nephropathy rats; 3. Sac group: DN rats + Sac group (orally, 60 mg/kg/d) for 6 weeks. After 6 weeks, the levels of serum albumin (ALB), glucose (GLU), creatinine (Cr), urea nitrogen (BUN) and 24-h urinary protein excretion were measured. In renal tissue homogenates, NLRP3 inflammasome, proinflammatory factors IL1-β and TNF-α, oxidative stress MDA and pro-fibrotic cytokine TGF-β1 were performed. Histological analysis of kidneys by hematoxylin and eosin (HE), PAS and Masson trichrome staining. RESULTS 1. Sacubitril/valsartan (Sac) significantly improved renal hypertrophy, proteinuria and serum albumin levels in rats with early diabetic nephropathy (P < 0.001), and decreased GLU, Scr (P<0.001), and BUN levels (P < 0.01).2. Light microscopy of renal tissues showed glomerular hypertrophy and interstitial inflammatory cell infiltration, and mean glomerular area (MGA) and mean glomerular volume (MGV) were crucially increased in early diabetic nephropathy (P < 0.001), and the Sac group showed reduced renal pathology and improved MGA and MGV (P < 0.001).3. Kidney tissue homogenate levels of NLRP3, Caspase-1, IL1-β, TNF-α, MDA and TGF-β1 were critically, increased in DN rats (P < 0.001), and SOD was significantly decreased. All these indicators above were improved after treatment (P < 0.0001). CONCLUSION Nlrp3-inflammasome promote progression of diabetic nephropathy through inflammation, fibrosis and oxidative stress; sacubitril/valsartan ameliorated early diabetes-induced renal damage by inhibiting NLRP3 pathway activation.
Collapse
Affiliation(s)
- Yan Pan
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu City, People’s Republic of China
- Correspondence: Yan Pan, Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Longzihu District, Bengbu City, Anhui Province, 233000, People’s Republic of China, Tel +86 13865030612, Email
| | - Lei Liu
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu City, People’s Republic of China
| | - Huijuan Yang
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu City, People’s Republic of China
| | - Weidong Chen
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu City, People’s Republic of China
| | - Zheng Chen
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu City, People’s Republic of China
| | - Jing Xu
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu City, People’s Republic of China
| |
Collapse
|
16
|
Fan X, Xu M, Chen X, Ren Q, Fan Y, Wang R, Chen J, Cui L, Wang Z, Sun X, Guo N. Proteomic profiling and correlations with clinical features reveal biomarkers indicative of diabetic retinopathy with diabetic kidney disease. Front Endocrinol (Lausanne) 2022; 13:1001391. [PMID: 36277688 PMCID: PMC9581084 DOI: 10.3389/fendo.2022.1001391] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 07/23/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022] Open
Abstract
Diabetic retinopathy (DR) and diabetic kidney disease (DKD) are complications of diabetes and place serious health and economic burdens on society. However, the identification and characterization of early biomarkers for DKD, especially for nonproliferative DR (NPDR) patients with DKD, are still needed. This study aimed to demonstrate the plasma proteomic profiles of NPDR+DKD and NPDR patients and identify potential biomarkers for early diagnosis of DKD. Fifteen plasma samples from the NPDR group and nine from the NPDR+DKD group were analyzed by LC-MS/MS to identify the differentially expressed proteins between the two groups. Functional enrichment, protein-protein interaction and clinical feature correlation analyses revealed the target protein candidates, which were verified using ELISA and receiver operating characteristic (ROC) analysis. In total, 410 proteins were detected in plasma; 15 were significantly upregulated and 7 were downregulated in the NPDR+DKD group. Bioinformatics analysis suggested that DKD is closely related to cell adhesion and immunity pathways. β-2-Microglobulin (B2M) and vimentin (VIM) were upregulated in NPDR+DKD, enriched as hub proteins and strongly correlated with clinical features. ELISA showed that B2M (p<0.001) and VIM (p<0.0001) were significantly upregulated in NPDR+DKD compared with NPDR. In ROC analysis, B2M and VIM could distinguish DKD from NPDR with area under the curve values of 0.9000 (p < 0.0001) and 0.9950. Our proteomic study revealed alterations in the proteomic profile and identified VIM and B2M as early biomarkers of DKD, laying the foundation for the prevention, diagnosis and treatment of DKD.
Collapse
Affiliation(s)
- Xiao’e Fan
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
- *Correspondence: Xiao’e Fan,
| | - Manhong Xu
- Department of Vitreoretinal and Ocular Trauma, Tianjin Medical University Eye Hospital, Eye Institute and School of Optometry, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin International Joint Research Center of Ophthalmology and Visual Science, Tianjin, China
| | - Xin Chen
- Department of Vitreoretinal and Ocular Trauma, Tianjin Medical University Eye Hospital, Eye Institute and School of Optometry, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin International Joint Research Center of Ophthalmology and Visual Science, Tianjin, China
| | - Qianfeng Ren
- Department of Pathology, Jincheng People’s Hospital, Jincheng, China
| | - Yan Fan
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Ranran Wang
- Department of Laboratory, Jincheng People’s Hospital, Jincheng, China
| | - Jiaqi Chen
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Li Cui
- Department of Nephrology, Jincheng People’s Hospital, Jincheng, China
| | - Zhengmin Wang
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Xiaoyan Sun
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| | - Nannan Guo
- Department of Ophthalmology, Jincheng People’s Hospital, Jincheng, China
| |
Collapse
|