1
|
Huang X, Chen L, He J, Tang J, Mou Z. Long non-coding RNA in IgA nephropathy: a comprehensive review. Ren Fail 2025; 47:2495836. [PMID: 40329456 PMCID: PMC12057784 DOI: 10.1080/0886022x.2025.2495836] [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: 07/30/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Immunoglobulin A nephropathy (IgAN) stands as the most prevalent primary glomerulonephritis globally, almost half of patients progress to end-stage kidney disease (ESKD). However, the precise pathogenesis of IgAN remains elusive. Long non-coding RNAs (lncRNAs), non-protein-coding transcripts that regulate gene expression, have been found to exhibit distinct expression patterns in various disease states. Comprehensive bioinformatic analyses from IgAN patients have uncovered differential expression of lncRNAs such as HOTAIR, H19, and MALAT1. Furthermore, a single nucleotide polymorphism in MIR31HG has been linked to IgAN susceptibility and correlated with clinical markers like urinary red blood cells and hemoglobin levels. Lnc-TSI and lnc-CHAF1B-3, specifically expressed in the kidneys of IgAN patients, exhibit associations with renal fibrosis indices and the degree of kidney function deterioration, influencing the progression of renal fibrosis through distinct signaling pathways. Additionally, renal intercellular adhesion molecule 1 (ICAM-1) related long noncoding RNA (ICR) levels positively correlate with IgAN severity and contribute to renal fibrosis, whereas serum H19 serves as an independent protective factor against IgAN. Notably, experiments have validated the involvement of PTTG3P, lnc-CHAF1B-3, and CRNDE in the pathogenesis of IgAN. Nevertheless, data on the roles of lncRNAs in IgAN pathogenesis and their potential as biomarkers remain limited, and effective therapeutic options for IgAN are similarly rare. Therefore, there is an urgent need to bridge this knowledge gap. This article presents a review of current literature on lncRNAs related to IgAN, aiming to consolidate existing findings and identify future research avenues.
Collapse
Affiliation(s)
- Xiaoxuan Huang
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lan Chen
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinxuan He
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jianhui Tang
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhixiang Mou
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
2
|
Tong Z, Zhao H, Cui C, Hong M, Ma Y, Sui L, Wang J, Yuan Q, Sun L. m6A-mediated regulation of ECA39 promotes renal fibrosis in chronic kidney disease by enhancing glycolysis and epithelial-mesenchymal transition. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119981. [PMID: 40315919 DOI: 10.1016/j.bbamcr.2025.119981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/24/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
Renal fibrosis is a vital pathological manifestation of chronic kidney disease (CKD). ECA39 is a conserved gene in the regulation of cell behavior; however, its function in renal fibrosis remains unclarified. A murine model of renal fibrosis was established by unilateral ureteral obstruction (UUO) operation. ECA39 expression was significantly upregulated in the kidneys of UUO mice. Prior to UUO operation (14 days), mice were administrated adeno-associated virus serotype 9 (AAV9, 1 × 1011 vector genomes) expressing ECA39 shRNA via tail vein injection. At postoperative day 7, AAV9-mediated inhibition of ECA39 was found to mitigate UUO-induced kidney damage, as manifested by reduced NGAL expression in kidneys, along with reduced serum creatinine and blood urea nitrogen (BUN) levels. Inhibition of ECA39 decreased collagen I, α-SMA and vimentin expression, but increased E-cadherin in kidney tissues. ECA39 inhibition reduced serum lactic acid level, increased ATP production, and suppressed glycolysis-related indicators HK2, PFKM, PKM2, PDK1, and LDHA expression. In parallel, human proximal tubular epithelial cells (HK-2) were treated with TGF-β1 (5 ng/ml, 48 h) to induce a cellular model of injury. ECA39 knockdown inhibit epithelial-mesenchymal transition (EMT) and glycolysis in HK-2 cells. Mechanistically, TGF-β1 treatment increased m6A modification of ECA39 mRNA, and the m6A "reader" IGF2BP2 knockdown reduced ECA39 mRNA stability. IGF2BP2 knockdown reduced lactic acid content and inhibited EMT in HK-2 cells, whereas ECA39 overexpression reversed these effects. Collectively, our studies demonstrated that inhibition of ECA39 suppresses glycolysis and EMT processes, thereby alleviating renal fibrosis in CKD.
Collapse
Affiliation(s)
- Ziyuan Tong
- Biological Therapy Department, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Hainan Zhao
- Department of Nephrology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, People's Republic of China
| | - Changwan Cui
- Biobank, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Mengqi Hong
- Department of Rehabilitation, Ningbo Ninth Hospital, Ningbo, Zhejiang, People's Republic of China
| | - Yutong Ma
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lu Sui
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jingyu Wang
- Renal Division, Peking University First Hospital, Beijing, People's Republic of China
| | - Quan Yuan
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Li Sun
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
3
|
Qian L, Hu W, Wang Y, Waheed YA, Hu S, Sun D, Li S. LncRNA TUG1 mitigates chronic kidney disease through miR-542-3p/HIF-1α/VEGF axis. Heliyon 2025; 11:e40891. [PMID: 39811365 PMCID: PMC11730199 DOI: 10.1016/j.heliyon.2024.e40891] [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: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Renal interstitial fibrosis (RIF) is a common pathway in chronic kidney disease (CKD) that ultimately leads to end-stage renal failure, worsening both glomerulosclerosis and interstitial fibrosis. Ten percent of the adult population in the world suffers from CKD, and as the ageing population continues to rise, it is increasingly regarded as a global threat-a silent epidemic. CKD has been discovered to be closely associated with both long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), while the precise molecular processes behind this relationship are still unclear. This study evaluated the impact of miR-542-3p and lncRNA TUG1 on renal fibrosis, along with the underlying regulatory mechanisms. Through in vitro tube formation assays, research demonstrated that knocking down lncRNA TUG1 may enhance angiogenesis and repair damaged endothelial cell-cell connections. We used Western blot and qRT-PCR methods in the unilateral ureteral obstruction (UUO) model to identify tissue hypoxia and fibrotic lesions. Additionally, a cutting-edge method known as fluorescence microangiography (FMA) was employed to detect damage to the peritubular capillaries (PTCs), with MATLAB software utilised for data evaluation. Furthermore, the coexpression of CD31 and α-SMA helped identify cells in the obstructed kidney that were transitioning from endothelium to myofibroblasts. On the contrary, lncRNA TUG1 downregulation showed a protective effect against the transition from endothelial cells to myofibroblasts. Additionally, knocking down lncRNA TUG1 has been shown to reduce the expression of fibrotic markers by alleviating tissue hypoxia. This effect was significantly counteracted by the inhibition of miR-542-3p. Collectively, our findings offer fresh perspectives on how lncRNA TUG1 and the miR-542-3p/HIF-1α/VEGF axis are regulated as renal fibrosis advances.
Collapse
Affiliation(s)
- Luoxiang Qian
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
- Department of Internal Medicine, Weinan Maternal and Child Health Hospital, Weinan, 714000, China
| | - Wanru Hu
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
| | - Yanping Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
| | | | - Shuqun Hu
- Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou, 221002, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China
| | - Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 221002, China
| |
Collapse
|
4
|
Wei Q, Huang J, Livingston MJ, Wang S, Dong G, Xu H, Zhou J, Dong Z. Pseudogene GSTM3P1 derived long non-coding RNA promotes ischemic acute kidney injury by target directed microRNA degradation of kidney-protective mir-668. Kidney Int 2024; 106:640-657. [PMID: 39074555 PMCID: PMC11416318 DOI: 10.1016/j.kint.2024.06.027] [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: 10/17/2023] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/31/2024]
Abstract
Long non-coding RNAs (lncRNAs) are a group of epigenetic regulators that have been implicated in kidney diseases including acute kidney injury (AKI). However, very little is known about the specific lncRNAs involved in AKI and the mechanisms underlying their pathologic roles. Here, we report a new lncRNA derived from the pseudogene GSTM3P1, which mediates ischemic AKI by interacting with and promoting the degradation of mir-668, a kidney-protective microRNA. GSTM3P1 and its mouse orthologue Gstm2-ps1 were induced by hypoxia in cultured kidney proximal tubular cells. In mouse kidneys, Gstm2-ps1 was significantly upregulated in proximal tubules at an early stage of ischemic AKI. This transient induction of Gstm2-ps1 depends on G3BP1, a key component in stress granules. GSTM3P1 overexpression increased kidney proximal tubular apoptosis after ATP depletion, which was rescued by mir-668. Notably, kidney proximal tubule-specific knockout of Gstm2-ps1 protected mice from ischemic AKI, as evidenced by improved kidney function, diminished tubular damage and apoptosis, and reduced kidney injury biomarker (NGAL) induction. To test the therapeutic potential, Gstm2-ps1 siRNAs were introduced into cultured mouse proximal tubular cells or administered to mice. In cultured cells, Gstm2-ps1 knockdown suppressed ATP depletion-associated apoptosis. In mice, Gstm2-ps1 knockdown ameliorated ischemic AKI. Mechanistically, both GSTM3P1 and Gstm2-ps1 possessed mir-668 binding sites and downregulated the mature form of mir-668. Specifically, GSTM3P1 directly bound to mature mir-668 to induce its decay via target-directed microRNA degradation. Thus, our results identify GSTM3P1 as a novel lncRNA that promotes kidney tubular cell death in AKI by binding mir-668 to inducing its degradation.
Collapse
Affiliation(s)
- Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.
| | - Jing Huang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Man Jiang Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Shixuan Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hongyan Xu
- Department of Biostatistics, Data Science and Epidemiology, School of Public Health, Augusta University, Augusta, Georgia, USA
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
5
|
Zhang C, Wang K, Chen X, Li Y. Mechanistic study on lncRNA XIST/miR-124-3p/ITGB1 axis in renal fibrosis in obstructive nephropathy. Exp Cell Res 2024; 442:114194. [PMID: 39127440 DOI: 10.1016/j.yexcr.2024.114194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/04/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE The purpose of this study was to investigate the role and possible mechanism of lncRNA XIST in renal fibrosis and to provide potential endogenous targets for renal fibrosis in obstructive nephropathy (ON). METHODS The study included 50 cases of ON with renal fibrosis (samples taken from patients undergoing nephrectomy due to ON) and 50 cases of normal renal tissue (samples taken from patients undergoing total or partial nephrectomy due to accidental injury, congenital malformations, and benign tumors). Treatment of human proximal renal tubular epithelium (HK-2) cells with TGF-β1 simulated renal fibrosis in vitro. Cell viability and proliferation were measured by CCK-8 and EdU, and cell migration was measured by transwell. XIST, miR-124-3p, ITGB1, and epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, α-SMA, and fibronectin) were detected by PCR and immunoblot. The targeting relationship between miR-124-3p and XIST or ITGB1 was verified by starBase and dual luciferase reporter gene experiments. In addition, The left ureter was ligated in mice as a model of unilateral ureteral obstruction (UUO), and the renal histopathology was observed by HE staining and Masson staining. RESULTS ON patients with renal fibrosis had elevated XIST and ITGB1 levels and reduced miR-124-3p levels. The administration of TGF-β1 exhibited a dose-dependent promotion of HK-2 cell viability, proliferation, migration, and EMT. Conversely, depleting XIST or enhancing miR-124-3p hindered HK-2 cell viability, proliferation, migration, and EMT in TGF-β1-damaged HK-2 cells HK-2 cells. XIST functioned as a miR-124-3p sponge. Additionally, miR-124-3p negatively regulated ITGB1 expression. Elevating ITGB1 weakened the impact of XIST depletion on TGF-β1-damaged HK-2 cells. Down-regulating XIST improved renal fibrosis in UUO mice. CONCLUSION XIST promotes renal fibrosis in ON by elevating miR-124-3p and reducing ITGB1 expressions.
Collapse
Affiliation(s)
- ChiTeng Zhang
- Department of Urology Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, 421001, China
| | - KangNing Wang
- Department of Urology Surgery, Xiangya Hospital Central South University, Changsha City, Hunan Province, 410000, China
| | - Xiang Chen
- Department of Urology Surgery, Xiangya Hospital Central South University, Changsha City, Hunan Province, 410000, China
| | - Yong Li
- Department of Urology Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, 421001, China.
| |
Collapse
|
6
|
Zhang G, Deng L, Jiang Z, Xiang G, Zeng Z, Zhang H, Wang Y. Titanium nanoparticles released from orthopedic implants induce muscle fibrosis via activation of SNAI2. J Nanobiotechnology 2024; 22:522. [PMID: 39215337 PMCID: PMC11363368 DOI: 10.1186/s12951-024-02762-4] [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] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Titanium alloys represent the prevailing material employed in orthopedic implants, which are present in millions of patients worldwide. The prolonged presence of these implants in the human body has raised concerns about possible health effects. This study presents a comprehensive analysis of titanium implants and surrounding tissue samples obtained from patients who underwent revision surgery for therapeutic reasons. The surface of the implants exhibited nano-scale corrosion defects, and nanoparticles were deposited in adjacent samples. In addition, muscle in close proximity to the implant showed clear evidence of fibrotic proliferation, with titanium content in the muscle tissue increasing the closer it was to the implant. Transcriptomics analysis revealed SNAI2 upregulation and activation of PI3K/AKT signaling. In vivo rodent and zebrafish models validated that titanium implant or nanoparticles exposure provoked collagen deposition and disorganized muscle structure. Snai2 knockdown significantly reduced implant-associated fibrosis in both rodent and zebrafish models. Cellular experiments demonstrated that titanium dioxide nanoparticles (TiO2 NPs) induced fibrotic gene expression at sub-cytotoxic doses, whereas Snai2 knockdown significantly reduced TiO2 NPs-induced fibrotic gene expression. The in vivo and in vitro experiments collectively demonstrated that Snai2 plays a pivotal role in mediating titanium-induced fibrosis. Overall, these findings indicate a significant release of titanium nanoparticles from the implants into the surrounding tissues, resulting in muscular fibrosis, partially through Snai2-dependent signaling.
Collapse
Affiliation(s)
- Gengming Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Linhua Deng
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhongjing Jiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Gang Xiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhuotong Zeng
- College of Environmental Science and Engineering, Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Hunan University, Ministry of Education, Changsha, 410082, PR China
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, 410011, PR China
| | - Hongqi Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yunjia Wang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
7
|
Liu H, Deng Y, Luo G, Yang Y, Xie B, Diao H, Chen M, Chen L, Xie P, Kwan HY, Zhao X, Sun X. DNA methylation of miR-181a-5p mediated by DNMT3b drives renal interstitial fibrosis developed from acute kidney injury. Epigenomics 2024; 16:945-960. [PMID: 39023272 PMCID: PMC11370974 DOI: 10.1080/17501911.2024.2370229] [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: 01/21/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Aim: To explore the role of miR-181a-5p in the progression of acute kidney injury (AKI) to renal interstitial fibrosis (RIF) from the perspective of DNA methylation.Materials & methods: The role of miR-181a-5p was confirmed by collecting clinical samples, injecting miR-181a-5p agomir into tail vein, and transfecting miR-181a-5p mimic in vitro. The mechanism of miR-181a-5p's influence on AKI induced RIF was investigated by methylation-specific PCR, bioinformatic analysis, transcriptome sequencing and so on.Results: MiR-181a-5p plays an important role in AKI induced RIF. DNMT3b-mediated miR-181a-5p promoter hypermethylation is the main reason for the downregulation of miR-181a-5p. HDAC9 and SNAI2 are direct targets of miR-181a-5p.Conclusion: Hypermethylation of miR-181a-5p promoter mediated by DNMT3b promotes AKI induced RIF by targeting HDAC9 and SNAI2.
Collapse
Affiliation(s)
- Huaxi Liu
- Department of Nephrology, TCM-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, 510315, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
- Boai Hospital of Zhongshan, Zhongshan, Guangdong528403, China
| | - Yijian Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Guanfeng Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Ying Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Bei Xie
- Department of Nephrology, TCM-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, 510315, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Huiling Diao
- Department of Nephrology, TCM-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, 510315, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Meilin Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Liqian Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Penghui Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoshan Zhao
- Department of Nephrology, TCM-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, 510315, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Xiaomin Sun
- Department of Nephrology, TCM-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, 510315, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong510515, China
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong510280, China
| |
Collapse
|
8
|
Zhao Y, Wang H, Tang Y, Wang J, Wu X, He Z, He Y, Tang Z. SNHG16/miR-205/HDAC5 is involved in the progression of renal fibrosis. J Biochem Mol Toxicol 2024; 38:e23617. [PMID: 38079211 DOI: 10.1002/jbt.23617] [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: 10/18/2022] [Revised: 10/31/2023] [Accepted: 11/28/2023] [Indexed: 01/18/2024]
Abstract
Renal interstitial fibrosis (RIF) represents an irreversible and progressive pathological manifestation of chronic renal disease, which ultimately leads to end-stage renal disease. Long noncoding RNAs (lncRNAs) have been suggested to be involved in the progression of RIF. Small nucleolar RNA host gene 16 (SNHG16), a member of lncRNAs, has been found to be involved in the progression of pulmonary fibrosis. This paper first researched the effect of SNHG16 on renal fibrosis. We established a unilateral ureteral obstruction (UUO)-induced mouse RIF model by ligation of the left ureter to evaluate the biological function of SNHG16 in RIF. As a result, SNHG16 was upregulated in UUO-induced renal fibrotic tissues. Knockdown of SNHG16 inhibited RIF and reduced alpha-smooth muscle actin (α-SMA), fibronectin, and college IV expression. miR-205 was a target of SNHG16, and downregulated in UUO-induced renal fibrotic tissues. Inhibition of miR-205 promoted RIF and increased the expression of α-SMA, college IV, and fibronectin. Overexpression of SNHG16 promoted the UUO-induced RIF, but miR-205 abrogated this effect of SNHG16. Histone deacetylase 5 (HDAC5) showed high expression in UUO-induced renal fibrotic tissues. Knockdown of HDAC5 significantly reduced α-SMA, fibronectin, and college IV expression in renal tissues of UUO-induced mice. Inhibition of miR-205 promoted HDAC5 expression, but knockdown of SNHG16 inhibited HDAC5 expression in renal tissues of UUO-induced mice. In conclusion, SHNG16 is highly expressed in renal fibrotic tissues of UUO-induced mice. Knockdown of SHNG16 may prevent UUO-induced RIF by indirectly upregulating HDAC5 via targeting miR-205. SHNG16 may be novel target for treating renal fibrosis.
Collapse
Affiliation(s)
- Yingdan Zhao
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Hanqing Wang
- Department of Nephrology, Jing'an District Center Hospital of Shanghai, Fudan University, Shanghai City, China
| | - Yunhai Tang
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Juan Wang
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Xia Wu
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Zifan He
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Yayun He
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Zhihuan Tang
- Department of Nephrology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| |
Collapse
|
9
|
Naas S, Schiffer M, Schödel J. Hypoxia and renal fibrosis. Am J Physiol Cell Physiol 2023; 325:C999-C1016. [PMID: 37661918 DOI: 10.1152/ajpcell.00201.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Renal fibrosis is the final stage of most progressive kidney diseases. Chronic kidney disease (CKD) is associated with high comorbidity and mortality. Thus, preventing fibrosis and thereby preserving kidney function increases the quality of life and prolongs the survival of patients with CKD. Many processes such as inflammation or metabolic stress modulate the progression of kidney fibrosis. Hypoxia has also been implicated in the pathogenesis of renal fibrosis, and oxygen sensing in the kidney is of outstanding importance for the body. The dysregulation of oxygen sensing in the diseased kidney is best exemplified by the loss of stimulation of erythropoietin production from interstitial cells in the fibrotic kidney despite anemia. Furthermore, hypoxia is present in acute or chronic kidney diseases and may affect all cell types present in the kidney including tubular and glomerular cells as well as resident immune cells. Pro- and antifibrotic effects of the transcription factors hypoxia-inducible factors 1 and 2 have been described in a plethora of animal models of acute and chronic kidney diseases, but recent advances in sequencing technologies now allow for novel and deeper insights into the role of hypoxia and its cell type-specific effects on the progression of renal fibrosis, especially in humans. Here, we review existing literature on how hypoxia impacts the development and progression of renal fibrosis.
Collapse
Affiliation(s)
- Stephanie Naas
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
10
|
Ni W, Zhou H, Lu H, Ma N, Hou B, Li W, Kong F, Yu J, Hou R, Jin J, Wen J, Zhang T, Meng X. Genetic and pharmacological inhibition of METTL3 alleviates renal fibrosis by reducing EVL m6A modification through an IGF2BP2-dependent mechanism. Clin Transl Med 2023; 13:e1359. [PMID: 37537731 PMCID: PMC10400756 DOI: 10.1002/ctm2.1359] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND N6 -methyladenosine (m6A) is of great importance in renal physiology and disease progression, but its function and mechanism in renal fibrosis remain to be comprehensively and extensively explored. Hence, this study will explore the function and potential mechanism of critical regulator-mediated m6A modification during renal fibrosis and thereby explore promising anti-renal fibrosis agents. METHODS Renal tissues from humans and mice as well as HK-2 cells were used as research subjects. The profiles of m6A modification and regulators in renal fibrosis were analysed at the protein and RNA levels using Western blotting, quantitative real-time polymerase chain reaction and other methods. Methylation RNA immunoprecipitation sequencing and RNA sequencing coupled with methyltransferase-like 3 (METTL3) conditional knockout were used to explore the function of METTL3 and potential targets. Gene silencing and overexpression combined with RNA immunoprecipitation were performed to investigate the underlying mechanism by which METTL3 regulates the Ena/VASP-like (EVL) m6A modification that promotes renal fibrosis. Molecular docking and virtual screening with in vitro and in vivo experiments were applied to screen promising traditional Chinese medicine (TCM) monomers and explore their mechanism of regulating the METTL3/EVL m6A axis and anti-renal fibrosis. RESULTS METTL3 and m6A modifications were hyperactivated in both the tubular region of fibrotic kidneys and HK-2 cells. Upregulated METTL3 enhanced the m6A modification of EVL mRNA to improve its stability and expression in an insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2)-dependent manner. Highly expressed EVL binding to Smad7 abrogated the Smad7-induced suppression of transforming growth factor-β (TGF-β1)/Smad3 signal transduction, which conversely facilitated renal fibrosis progression. Molecular docking and virtual screening based on the structure of METTL3 identified a TCM monomer named isoforsythiaside, which inhibited METTL3 activity together with the METTL3/EVL m6A axis to exert anti-renal fibrosis effects. CONCLUSIONS Collectively, the overactivated METTL3/EVL m6A axis is a potential target for renal fibrosis therapy, and the pharmacological inhibition of METTL3 activity by isoforsythiaside suggests that it is a promising anti-renal fibrosis agent.
Collapse
Affiliation(s)
- Wei‐Jian Ni
- Department of PharmacyAnhui Provincial Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiPeople's Republic of China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Hong Zhou
- Department of PharmacyAnhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiPeople's Republic of China
| | - Hao Lu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Nan‐Nan Ma
- Department of UrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Bing‐Bing Hou
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Wei Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Fan‐Xu Kong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
- Department of PharmacyThe Second People's Hospital of HefeiHefeiAnhuiPeople's Republic of China
| | - Ju‐Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Rui Hou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Juan Jin
- Research Center for Translational MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
- School of Basic MedicineAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Jia‐Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Tao Zhang
- Department of UrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiPeople's Republic of China
| | - Xiao‐Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Institute of Innovative DrugsSchool of PharmacyAnhui Medical UniversityHefeiAnhuiPeople's Republic of China
| |
Collapse
|
11
|
Du J, Zhang X, Zhang J, Huo S, Li B, Wang Q, Song M, Shao B, Li Y. Necroptosis and NLPR3 inflammasome activation mediated by ROS/JNK pathway participate in AlCl 3-induced kidney damage. Food Chem Toxicol 2023; 178:113915. [PMID: 37393014 DOI: 10.1016/j.fct.2023.113915] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
Aluminum (Al) is a common environmental pollutant that can induce kidney damage. However, the mechanism is not clear. In the present study, to explored the exact mechanism of AlCl3-induced nephrotoxicity, C57BL/6 N male mice and HK-2 cells were used as experimental subjects. Our results showed that Al induced reactive oxygen species (ROS) overproduction, c-Jun N-terminal kinase (JNK) signaling activation, RIPK3-dependent necroptosis, NLRP3 inflammasome activation, and kidney damage. In addition, inhibiting JNK signaling could downregulate the protein expressions of necroptosis and NLRP3 inflammasome, thereby alleviating kidney damage. Meanwhile, clearing ROS effectively inhibited JNK signaling activation, which in turn inhibited necroptosis and NLRP3 inflammasome activation, ultimately alleviating kidney damage. In conclusion, these findings suggest that necroptosis and NLPR3 inflammasome activation mediated by ROS/JNK pathway participate in AlCl3-induced kidney damage.
Collapse
Affiliation(s)
- Jiayu Du
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xuliang Zhang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jian Zhang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Siming Huo
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bo Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Qi Wang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Miao Song
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bing Shao
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
12
|
Sen I, Uchida S, Garikipati VNS. Long non-coding RNA lnc-CHAF1B-3 as a new player in fibrosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:566-567. [PMID: 36910715 PMCID: PMC9996120 DOI: 10.1016/j.omtn.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
- Ilayda Sen
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Frederikskaj 10B, 2. (building C), 2450 Copenhagen SV, Denmark
| | - Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.,Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|