1
|
Liu Y, Wu R, Zhou Z, Zhou J, Zhang J, Wang X. Combining mitochondrial proteomes and Mendelian randomization to identify novel therapeutic targets for diabetic nephropathy. Ren Fail 2025; 47:2473669. [PMID: 40125968 PMCID: PMC11934170 DOI: 10.1080/0886022x.2025.2473669] [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: 01/02/2025] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/25/2025] Open
Abstract
Diabetic nephropathy (DN) is a common microvascular complication of diabetes. Mitochondrial dysfunction in the kidney caused by diabetes has previously been linked to the pathogenesis of DN. By mass spectrometry, we identified characteristic proteins of DN from the renal mitochondria in mouse model. To identify the core proteins among them, Mendelian randomization (MR) analysis, microarray data validation, and drug-target interaction analysis were employed. MR analysis found that 189 candidate targets had a causal link with DN risk factors (estimated glomerular filtration rate (eGFR), urinary albumin excretion, and serum creatinine). After systematic analysis, we validated that SLC25A16, CTNND1, C2CD2L, ALDH3A2, NEU1, APEH, CORO1A, NUDT19, and NDUFA4L2 are the core proteins with promising druggability in DN. This study suggests the feasibility of using MR analysis for DN drug target screening, and provides potential insights into mitochondrial dysfunction research, which may contribute to further DN pathogenesis exploration.
Collapse
Affiliation(s)
- Yang Liu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Huzhou Key Laboratory of Chronic Kidney Disease, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Rong Wu
- Huzhou Key Laboratory of Chronic Kidney Disease, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Zhelun Zhou
- Huzhou Key Laboratory of Chronic Kidney Disease, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Junan Zhou
- Huzhou Key Laboratory of Chronic Kidney Disease, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Jiaai Zhang
- Huzhou Key Laboratory of Chronic Kidney Disease, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Xiaoyi Wang
- Huzhou Key Laboratory of Chronic Kidney Disease, First Affiliated Hospital of Huzhou University, Huzhou, China
| |
Collapse
|
2
|
Yu X, Hu Y, Jiang W. Integrative analysis of mitochondrial and immune pathways in diabetic kidney disease: identification of AASS and CASP3 as key predictors and therapeutic targets. Ren Fail 2025; 47:2465811. [PMID: 39988817 PMCID: PMC11852243 DOI: 10.1080/0886022x.2025.2465811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVES Diabetic kidney disease (DKD) is driven by mitochondrial dysfunction and immune dysregulation, yet the mechanistic interplay remains poorly defined. This study aimed to identify key molecular networks linking mitochondrial and immune pathways to DKD progression, with a focus on uncovering biomarkers and therapeutic targets. METHODS We conducted an integrative analysis of human DKD cohorts (GSE30122, GSE96804) using weighted gene co-expression network analysis (WGCNA) to identify gene modules enriched for immune response genes and mitochondrial pathways (from MitoCarta3.0). Machine learning algorithms were employed to prioritize key biomarkers for further investigation. Experimental validation was performed using a DKD rat model. RESULTS WGCNA revealed significant gene modules associated with immune responses and mitochondrial functions. Machine learning analysis highlighted two central biomarkers: aminoadipate-semialdehyde synthase (AASS) and caspase-3 (CASP3). In the DKD rat model, elevated levels of AASS and CASP3 were found to correlate with increased oxidative stress. Mechanistically, AASS was shown to drive mitochondrial damage via lysine metabolism, while CASP3 amplified inflammatory apoptosis pathways. CONCLUSIONS Our findings establish AASS and CASP3 as dual biomarkers and therapeutic targets, bridging mitochondrial-immune crosstalk to DKD pathogenesis. This multi-omics framework provides actionable insights for targeting kidney damage in diabetes.
Collapse
Affiliation(s)
- Xinxin Yu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Nephrology, Qingdao Eighth People’s Hospital, Qingdao, Shandong, China
| | - Yongzheng Hu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei Jiang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
3
|
Wang G, Mu W, Zhang T. Comment on 'the relationship between kidney disease and mitochondria: a bibliometric study'. Ren Fail 2025; 47:2460747. [PMID: 39995106 PMCID: PMC11864025 DOI: 10.1080/0886022x.2025.2460747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Affiliation(s)
- Guozhen Wang
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Wenwen Mu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Tianxing Zhang
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| |
Collapse
|
4
|
Pei X, Liu J, Wei Y, Zhu Y, Li Y, Wang Q, Zhuang L, Jin G. PINK1 affects mitochondrial oxidative phosphorylation by regulating MFN2 to alleviate diabetic kidney disease. Int Immunopharmacol 2025; 157:114786. [PMID: 40319746 DOI: 10.1016/j.intimp.2025.114786] [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: 02/01/2025] [Revised: 04/17/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is widely recognized as a prevalent and major microvascular complication of diabetes mellitus. Mitofusin 2 (MFN2) has been closely linked to the development of diabetes mellitus, yet its precise role in the pathogenesis of DKD remains uncertain. The objective of our current research was to explore the role of MFN2 in the advanced DKD and its underlying molecular pathway. This research was to examine the involvement and molecular pathways of MFN2 in the advancement of DKD. METHODS In this study, MFN2 was manipulated in high glucose (HG)-treated HK2 cells to investigate its impact on cell proliferation, apoptosis, and mitochondrial oxidative phosphorylation. Models of MFN2 overexpression or silencing were established in db/db mice as a diabetes model. The alterations in kidney morphology, renal fibrosis severity, macrophage polarization, and related inflammatory factors (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]) were evaluated. Furthermore, HK2 cells were co-cultivated with M1-type macrophages to examine the impact of MFN2 expression on macrophage polarization. Subsequently, we delved deeper into the upstream mechanisms utilizing the STRING database. RESULTS Our study identified that MFN2 expression was downregulated in HG-treated HK-2 cells. The overexpression of MFN2 resulted in increased HK2 cell proliferation, improved degree of oxidative phosphorylation, and reduced apoptosis. In db/db mice, MFN2 overexpression exerted a protective effect on DKD-induced nephropathy and fibrosis. Notably, MFN2 overexpression influenced macrophage polarization and modulated expression of related inflammatory factors by promoting mitochondrial oxidative phosphorylation. Additionally, STRING database prediction revealed that PTEN-induced kinase 1 (PINK1) regulated MFN2 expression, which was consistent with MFN2 changes in DKD, and this regulation was associated with DKD progression. CONCLUSION The role of MFN2 in maintaining mitochondrial function in DKD may be regulated by PINK1 and provides a new potential therapeutic target for DKD.
Collapse
Affiliation(s)
- Xiaoyan Pei
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China.
| | - Jie Liu
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Yu Wei
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Yanhui Zhu
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Yu Li
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Qiong Wang
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Langen Zhuang
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China
| | - Guoxi Jin
- Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, PR China.
| |
Collapse
|
5
|
Li XQ, Xiao ZZ, Ma K, Liu XY, Liu HH, Hu B, Zhao Q, Li HY, Chen RC, Meng Y, Yin LH. Angiotensin-Converting Enzyme-Dependent Intrarenal Angiotensin II Contributes to CTP: Phosphoethanolamine Cytidylyltransferase Downregulation, Mitochondrial Membranous Disruption, and Reactive Oxygen Species Overgeneration in Diabetic Tubulopathy. Antioxid Redox Signal 2025; 42:767-786. [PMID: 39495586 DOI: 10.1089/ars.2024.0637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
Aims: The limited therapeutic options for diabetic tubulopathy (DT) in early diabetic kidney disease (DKD) reflect the difficulty of targeting renal tubular compartment. While renin-angiotensin-aldosterone system (RAS) inhibitors are commonly utilized in the management of DKD, how intrarenal RAS contributes to diabetic tubular injury is not fully understood. Mitochondrial disruption and reactive oxygen species (ROS) overgeneration have been involved in diabetic tubular injury. Herein, we aim to test the hypothesis that angiotensin-converting enzyme (ACE)-dependent intrarenal angiotensin II (AngII) disrupts tubular mitochondrial membranous homeostasis and causes excessive ROS generation in DT. Results: Mice suffered from renal tubular mitochondrial disruption and ROS overgeneration following high-fat diet/streptozocin-type 2 diabetic induction. Intrarenal AngII generation is ACE-dependent in DT. Local AngII accumulation in renal tissues was achieved by intrarenal artery injection. ACE-dependent intrarenal AngII-treated mice exhibit markedly elevated levels of makers of tubular injury. CTP: Phosphoethanolamine cytidylyltransferase (PCYT2), the primary regulatory enzyme for the biosynthesis of phosphatidylethanolamine, was enriched in renal tubules according to single-cell RNA sequencing. ACE-dependent intrarenal AngII-induced tubular membranous disruption, ROS overgeneration, and PCYT2 downregulation. The diabetic ambiance deteriorated the detrimental effect of ACE-dependent intrarenal AngII on renal tubules. Captopril, the ACE inhibitor (ACEI), showed efficiency in partially ameliorating ACE-dependent intrarenal AngII-induced tubular deterioration pre- and post-diabetic induction. Innovation and Conclusion: This study uncovers a critical role of ACE-dependent intrarenal AngII in mitochondrial membranous disruption, ROS overgeneration, and PCYT2 deficiency in diabetic renal tubules, providing novel insight into DT pathogenesis and ACEI-combined therapeutic targets. Antioxid. Redox Signal. 42, 767-786.
Collapse
Affiliation(s)
- Xia-Qing Li
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Nephrology Department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Zhang-Zhang Xiao
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Nephrology Department, Houjie Hospital of Dongguan, Dongguan, China
| | - Ke Ma
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Nephrology Department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Xia-Yun Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Nephrology Department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Huan-Huan Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Bo Hu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Qian Zhao
- Department of Infectious Diseases and Hepatology Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong-Yue Li
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Nephrology Department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Rui-Chang Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Yu Meng
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Nephrology Department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Liang-Hong Yin
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Huangpu Institute of Materials, Guangzhou, China
| |
Collapse
|
6
|
Feng A, Yin R, Xu R, Zhang B, Yang L. An update on renal tubular injury as related to glycolipid metabolism in diabetic kidney disease. Front Pharmacol 2025; 16:1559026. [PMID: 40303925 PMCID: PMC12038058 DOI: 10.3389/fphar.2025.1559026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes, which can result in end-stage renal disease (ESRD). As the main site of renal reabsorption and its exposed environment, renal tubules can be damaged by various factors. Recent studies have shown that renal tubular epithelial cells (RTECs) injury plays an important role in the occurrence and progression of DKD. The glycolipid metabolism disorders are a vital factor contributing to RTECs injury, which in turn affects the progression of DKD. Abnormal glucose and lipid metabolism can cause oxidative stress, mitochondrial damage, cell apoptosis and lipid accumulation, which can cause RTECs injury. Therefore, this review describes the main pathological mechanism of the injury caused by glycolipid metabolism and the corresponding therapeutic drugs in the clinical treatment of DKD.
Collapse
Affiliation(s)
- Anqi Feng
- Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Prevention and Research Care, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Ruili Yin
- Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Prevention and Research Care, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Rong Xu
- Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Prevention and Research Care, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Baoyu Zhang
- Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Prevention and Research Care, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Longyan Yang
- Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Prevention and Research Care, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Gong YX. Research progress of effective components of traditional Chinese medicine in intervening apoptosis of renal tubular epithelial cells in diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119874. [PMID: 40280372 DOI: 10.1016/j.jep.2025.119874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Apoptosis of renal tubular epithelial cells (RTECs) is a critical pathological feature of diabetic kidney disease (DKD), a primary contributor to end-stage renal disease (ESRD). Traditional Chinese medicine (TCM) has shown potential in modulating RTECs apoptosis and alleviating DKD progression, making it a promising area for further investigation. AIM OF THE STUDY This study aims to summarize the apoptotic pathways implicated in DKD, analyze existing research on the effects of TCM monomers and compounds on RTECs apoptosis, and elucidate the molecular mechanisms underlying these effects. Additionally, this study emphasizes the significant role of TCM in mitigating DKD progression. MATERIALS AND METHODS Relevant literature was systematically retrieved from ancient Chinese medicine texts and modern scientific databases, including CNKI, Web of Science, and PubMed, using keywords such as "Traditional Chinese Medicine", "Diabetic Kidney Disease", "Diabetic Nephropathy", "Renal Tubular Epithelial Cells", and "Apoptosis". The collected information was synthesized and analyzed. RESULTS This review systematically analyzed 187 relevant studies, focusing on the mechanisms and clinical applications of 16 TCM monomers and 20 TCM compounds in DKD treatment. Key bioactive compounds, such as berberine, astragaloside IV, and tanshinone IIA, have demonstrated renoprotective effects by mitigating oxidative stress and inflammation, as well as regulating critical signaling pathways, including PI3K/Akt, NF-κB, and TGF-β/Smad, to suppress RTECs apoptosis and decelerate DKD progression. Additionally, several TCM compounds have shown significant efficacy in clinical studies, reducing proteinuria and enhancing renal function, thereby reinforcing the therapeutic potential of TCM in DKD management. CONCLUSIONS RTECs apoptosis is a critical pathological feature of DKD. TCM exhibits significant therapeutic potential by intervening in this process through multiple pathways. This study highlights the ability of TCM to exert anti-apoptotic and renoprotective effects by modulating oxidative stress, inflammatory responses, and multiple cellular signaling pathways. The multi-component and multi-target characteristics of TCM offer a promising avenue for the development of novel therapeutic strategies. However, further rigorous research and high-quality clinical trials are required to validate its efficacy and elucidate its mechanisms of action.
Collapse
Affiliation(s)
- Yu Xin Gong
- Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
8
|
Shan XM, Lu C, Chen CW, Wang CT, Liu TT, An T, Zhu ZY, Zou DW, Gao YB. Tangshenning formula alleviates tubular injury in diabetic kidney disease via the Sestrin2/AMPK/PGC-1α axis: Restoration of mitochondrial function and inhibition of ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119579. [PMID: 40043828 DOI: 10.1016/j.jep.2025.119579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tangshenning (TSN) is a traditional Chinese medicinal formula developed on principles of kidney tonification and collateral unblocking. TSN, formulated from Astragalus mongholicus Bunge, Rheum palmatum L., Ligusticum chuanxiong Hort., and Rosa laevigata Michx., has demonstrated significant clinical efficacy in the treatment of diabetic kidney disease (DKD). Our previous studies have suggested that TSN mitigates tubular injury in DKD by inhibiting ferroptosis, however, the precise molecular targets and mechanistic pathways underlying these effects remain to be fully elucidated. AIM OF THE STUDY We investigated whether the Sestrin2/AMPK/PGC-1α axis serves as a key pathway mediating TSN's protective effects against tubular injury in DKD. METHODS In vivo, a spontaneous DKD mouse model was developed using KK-Ay mice. In vitro, human tubular epithelial cells (TECs) were used to establish high glucose and ferroptosis models, as well as a Sestrin2 knockdown model for further analysis. Molecular docking was utilized to examine the binding interactions between TSN's key active components and Sestrin2. Colocalization of Sestrin2 and GPX4 was assessed using dual fluorescence staining. Protein expression levels related to the Sestrin2/AMPK/PGC-1α pathway, ferroptosis markers (SLC7A11 and GPX4), and the tubular injury marker KIM-1 were quantified via Western blot analysis. In vivo, DHE staining, TUNEL staining, and ferrous ion content measurement were performed to evaluate ferroptosis levels in renal tissue. In vitro, the BODIPY 581/591 C11 probe and ferrous ion assay were used to assess ferroptosis levels in TECs. MitoSOX staining, JC-1 assay, and ATP level measurements were conducted to evaluate mitochondrial function in TECs. RESULTS In vivo, our results demonstrated that TSN improved renal function, alleviated tubular injury, and reduced pathological damage in DKD mice. Furthermore, TSN upregulated the protein expression of the Sestrin2/AMPK/PGC-1α axis and decreased ferroptosis-related markers in the DKD mouse model. Similarly, in vitro, TSN enhanced the expression of the Sestrin2/AMPK/PGC-1α pathway, restored mitochondrial function, and inhibited ferroptosis in TECs under high glucose and ferroptosis-inducing conditions. Additionally, downregulation of Sestrin2 impaired the therapeutic effects of TSN. CONCLUSION TSN alleviates tubular injury in DKD by activating the Sestrin2/AMPK/PGC-1α pathway, restoring mitochondrial function, and inhibiting ferroptosis in TECs.
Collapse
Affiliation(s)
- Xiao-Meng Shan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Cong Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Chun-Wei Chen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Cui-Ting Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Tian-Tian Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Tian An
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Zhi-Yao Zhu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China
| | - Da-Wei Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China.
| | - Yan-Bin Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, People's Republic of China.
| |
Collapse
|
9
|
Fang H, Sun X, Ding Y, Niu B, Chen Q. Loureirin B analogs mitigate oxidative stress and confer renal protection. Cell Signal 2025; 132:111787. [PMID: 40188928 DOI: 10.1016/j.cellsig.2025.111787] [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: 01/09/2025] [Revised: 03/13/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Diabetic kidney disease (DKD) is a microvascular complication of diabetes with high morbidity and mortality, necessitating effective treatment. In this study, the Loureirin B analogue (LB-A) was utilized to treat DKD in mice. The results demonstrated that LB-A effectively prevent the progression of DKD in mice, significantly lowering fasting blood glucose levels and reducing proteinuria levels. Additionally, there was a significant decrease in oxidase content in the kidneys of mice, accompanied by an increase in antioxidant oxidase content, resulting in a decrease in ROS levels, mitigating oxidative stress state through modulation of Cxcl1. Cell experiments further confirmed that reducing Cxcl1/Cxcr2 axis activation prevented the onset of DKD induced by high glucose exposure and affected the therapeutic effect of LB-A as well. These findings provide evidences to support that LB-A may mitigate oxidative stress by modulating the Cxcl1 signaling pathway, thereby contributing to renal protection in the context of DKD treatment.
Collapse
Affiliation(s)
- Haowen Fang
- School of environmental and chemical engineering, Shanghai University, Shanghai, PR China.
| | - Xiaodong Sun
- Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Medicine, Shanghai University, Shanghai, PR China.
| | - Yanting Ding
- Tongji Hospital, School of Medicine, Tongji University, Shanghai, PR China; Shanghai Biochip Co., Ltd., National Engineering Center for Biochip at Shanghai, Shanghai, PR China.
| | - Bing Niu
- School of Life Sciences, Shanghai University, Shanghai, PR China.
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai, PR China.
| |
Collapse
|
10
|
Li Q, Shang J, Inagi R. Control of Mitochondrial Quality: A Promising Target for Diabetic Kidney Disease Treatment. Kidney Int Rep 2025; 10:994-1010. [PMID: 40303215 PMCID: PMC12034889 DOI: 10.1016/j.ekir.2024.12.029] [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: 07/29/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 05/02/2025] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD), affecting over 40% of patients with diabetes. DKD progression involves fibrosis and damage to glomerular and tubulointerstitial regions, with mitochondrial dysfunction playing a critical role. Impaired mitochondria lead to reduced adenosine triphosphate (ATP) production, damaged mitochondria accumulation, and increased reactive oxygen species (ROS), contributing to renal deterioration. Maintaining mitochondrial quality control (MQC) is essential for preventing cell death, tissue injury, and kidney failure. Recent clinical trials show that enhancing MQC can alleviate DKD. However, current treatments cannot halt kidney function decline, underscoring the need for new therapeutic strategies. Mitochondrial-targeted drugs show potential; however, challenges remain because of adverse effects and unclear mechanisms. Future research should aim to comprehensively explore therapeutic potential of MQC in DKD. This review highlights the significance of MQC in DKD treatment, emphasizing the need to maintain mitochondrial quality for developing new therapies.
Collapse
Affiliation(s)
- Qi Li
- Division of Chronic Kidney Disease Pathophysiology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Jin Shang
- Division of Chronic Kidney Disease Pathophysiology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Zeinali Nia E, Najjar Sadeghi R, Ebadi M, Faghihi M. ERK1/2 gene expression and hypomethylation of Alu and LINE1 elements in patients with type 2 diabetes with and without cataract: Impact of hyperglycemia-induced oxidative stress. J Diabetes Investig 2025; 16:689-706. [PMID: 39804191 PMCID: PMC11970314 DOI: 10.1111/jdi.14405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 04/05/2025] Open
Abstract
AIMS This study aimed to delineate the effect of hyperglycemia on the Alu/LINE-1 hypomethylation and in ERK1/2 genes expression in type 2 diabetes with and without cataract. METHODS This study included 58 diabetic patients without cataracts, 50 diabetic patients with cataracts, and 36 healthy controls. After DNA extraction and bisulfite treatment, LINE-1 and Alu methylation levels were assessed using Real-time MSP. ERK1/2 gene expression was analyzed through real-time PCR. Total antioxidant capacity (TAC), and fasting plasma glucose (FPG) were measured using colorimetric methods. Statistical analysis was performed with SPSS23, setting the significance level at P < 0.05. RESULTS The TAC levels were significantly lower for cataract and diabetic groups than controls (259.31 ± 122.99, 312.43 ± 145.46, 372.58 ± 132.95 nanomole of Trolox equivalent) with a significant correlation between FPG and TAC levels in both the cataract and diabetic groups (P < 0.05). Alu and LINE-1 sequences were found to be statistically hypomethylated in diabetic and cataract patients compared to controls. In these groups, TAC levels were directly correlated with Alu methylation (P < 0.05) but not LINE-1. ERK1/2 gene expression was significantly higher in diabetic and cataract patients, showing increases of 2.41-fold and 1.43-fold for ERK1, and 1.27-fold and 1.5 for ERK2, respectively. ERK1 expression correlated significantly with FPG levels. A reverse correlation was observed between TAC levels and ERK1/2 expression. CONCLUSIONS Our findings indicate that hyperglycemia-induced oxidative stress may alter ERK1/2 gene expression patterns and induce aberrant hypomethylation in Alu and LINE-1 sequences. These aberrant changes may play a contributing role in diabetic complications such as cataracts.
Collapse
Affiliation(s)
- Elham Zeinali Nia
- Department of Biochemistry, Faculty of Basic SciencesIslamic Azad University Damghan BranchDamghanIran
| | - Ruhollah Najjar Sadeghi
- Department of Clinical Biochemistry, Faculty of MedicineMazandaran University of Medical SciencesSariIran
| | - Mostafa Ebadi
- Department of Biochemistry, Faculty of Basic SciencesIslamic Azad University Damghan BranchDamghanIran
| | - Mohammad Faghihi
- Department of Medical SciencesShahid Beheshti UniversityTehranIran
| |
Collapse
|
12
|
Liu J, Feng L, Jia Q, Meng J, Zhao Y, Ren L, Yan Z, Wang M, Qin J. A comprehensive bioinformatics analysis identifies mitophagy biomarkers and potential Molecular mechanisms in hypertensive nephropathy. J Biomol Struct Dyn 2025; 43:3204-3223. [PMID: 38334110 DOI: 10.1080/07391102.2024.2311344] [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: 07/31/2023] [Accepted: 12/05/2023] [Indexed: 02/10/2024]
Abstract
Mitophagy, the selective removal of damaged mitochondria, plays a critical role in kidney diseases, but its involvement in hypertensive nephropathy (HTN) is not well understood. To address this gap, we investigated mitophagy-related genes in HTN, identifying potential biomarkers for diagnosis and treatment. Transcriptome datasets from the Gene Expression Omnibus database were analyzed, resulting in the identification of seven mitophagy related differentially expressed genes (MR-DEGs), namely PINK1, ULK1, SQSTM1, ATG5, ATG12, MFN2, and UBA52. Further, we explored the correlation between MR-DEGs, immune cells, and inflammatory factors. The identified genes demonstrated a strong correlation with Mast cells, T-cells, TGFβ3, IL13, and CSF3. Machine learning techniques were employed to screen important genes, construct diagnostic models, and evaluate their accuracy. Consensus clustering divided the HTN patients into two mitophagy subgroups, with Subgroup 2 showing higher levels of immune cell infiltration and inflammatory factors. The functions of their proteins primarily involve complement, coagulation, lipids, and vascular smooth muscle contraction. Single-cell RNA sequencing revealed that mitophagy was most significant in proximal tubule cells (PTC) in HTN patients. Pseudotime analysis of PTC confirmed the expression changes observed in the transcriptome. Intercellular communication analysis suggested that mitophagy might regulate PTC's participation in intercellular crosstalk. Notably, specific transcription factors such as HNF4A, PPARA, and STAT3 showed strong correlations with mitophagy-related genes in PTC, indicating their potential role in modulating PTC function and influencing the onset and progression of HTN. This study offers a comprehensive analysis of mitophagy in HTN, enhancing our understanding of the pathogenesis, diagnosis, and treatment of HTN.
Collapse
Affiliation(s)
- Jiayou Liu
- The Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Luda Feng
- Department of Nephropathy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Jia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Meng
- Department of Nephropathy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yun Zhao
- Department of Nephropathy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Ren
- Department of Nephropathy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ziming Yan
- Department of Nephropathy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Manrui Wang
- The Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jianguo Qin
- Department of Nephropathy, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Fan G, Tian M, Pan H, Sun C, Gong H, Luo Q, Yuan J. Label-free whole-kidney metabolic optical imaging using in vivo insulated cryofixation and cryo-micro-optical sectioning tomography. BIOMEDICAL OPTICS EXPRESS 2025; 16:1513-1527. [PMID: 40322009 PMCID: PMC12047708 DOI: 10.1364/boe.554000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 05/08/2025]
Abstract
Cryo-imaging has the potential to obtain and visualize the metabolic state of the whole kidney without labeling. However, uneven fixation of metabolic information and incomplete organ morphology in three dimensions limit cryo-imaging application. Here, a pipeline of in vivo insulated cryofixation combined with cryo-micro optical sectioning tomography (cryo-MOST) was established to achieve uniform and complete cryofixation and three-dimensional visualization of renal metabolic mapping at a micron-scale resolution. By this pipeline, we discovered an increased renal redox ratio of db/db mice with type 2 diabetes, indicating the presence of metabolic disorders. The results demonstrate that our convenient optical imaging tool provides a micro-resolution, quantitative assessment of the metabolic state of the whole kidney and potentially extends to other organs.
Collapse
Affiliation(s)
- Guoqing Fan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- MOE Key Laboratory for Biomedical Photonics, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingyu Tian
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- MOE Key Laboratory for Biomedical Photonics, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Huijuan Pan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- MOE Key Laboratory for Biomedical Photonics, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chuancao Sun
- Institute of Future Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haoyu Gong
- MOE Key Laboratory for Biomedical Photonics, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- MOE Key Laboratory for Biomedical Photonics, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, Suzhou 215123, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
- MOE Key Laboratory for Biomedical Photonics, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, Suzhou 215123, China
| |
Collapse
|
14
|
Yang R, Liu W, Zhou Y, Cheng B, Liu S, Wu R, Liu Y, Li J. Modulating HIF-1α/HIF-2α homeostasis with Shen-Qi-Huo-Xue formula alleviates tubular ferroptosis and epithelial-mesenchymal transition in diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119478. [PMID: 39947365 DOI: 10.1016/j.jep.2025.119478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetic kidney disease (DKD) is one of the main types of chronic kidney disease, which seriously affects the quality of life of patients. Shen-Qi-Huo-Xue formula (SQHXF), based on the Shen-Qi-Di-Huang decoction, is a traditional Chinese medicine formula for DKD. This study explored the mechanism of action of SQHXF on DKD through analysis of drug components, in vivo and in vitro experiments. AIM OF THE STUDY To elucidate the regulatory mechanisms of HIF-1α/HIF-2α homeostasis on ferroptosis and epithelial-mesenchymal transition (EMT) in renal tubular epithelial cells and the mechanism of action of SQHXF against DKD. METHODS The components of SQHXF were analyzed using UPLC-Q Exactive HF/MS. The effects of SQHXF on renal function, urinary proteins, glucose-lipid metabolism, hepatic function, renal tissue hypoxia, ferroptosis and EMT were analyzed following gavage of DKD model mice with different SQHXF doses. The effects of changes in HIF-1α and HIF-2α expression on ferroptosis and EMT, as well as the modulatory effects of SQHXF-containing serum, were assessed in vitro. The potential feedback mechanism of HIFs/ferroptosis/EMT was elucidated using HIF-1α knockdown and a ferroptosis inhibitor. RESULTS One-hundred and fifty compounds in SQHXF were tested for bloodstream entry. In vivo study showed that SQHXF was able to reduce creatinine, uric acid, fasting plasma glucose, 24-h urinary protein, low-density lipoprotein cholesterol, and aspartate aminotransferase levels, up-regulate HIF-1α, down-regulate HIF-2α, reduce ferroptosis, and alleviate renal fibrosis and EMT in tubular epithelial cells. HIF-1α/HIF-2α imbalance promoted ferroptosis and EMT in HK-2 cells, which was attenuated by SQHXF-containing serum. HIF-1α knockdown decreased HIF-2α expression and reduced ferroptosis and EMT. Inhibition of ferroptosis reduced EMT but failed to regulate HIF-1α and HIF-2α. CONCLUSIONS SQHXF alleviated ferroptosis and EMT, improved liver and kidney function, reduced proteinuria, and alleviated renal lesions by maintaining equilibrium between HIF-1α and HIF-2α.
Collapse
Affiliation(s)
- Ronglu Yang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Wu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Yi Zhou
- Graduate School of Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Bin Cheng
- Outpatient Department, Anhui University of Traditional Chinese Medicine, Hefei, 230031, China.
| | - Shiyi Liu
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Ruiying Wu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Yongjun Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Jinhu Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
15
|
Yang T, Peng Y, Shao Y, Pan D, Cheng Q, Jiang Z, Qian S, Li B, Yan M, Zhu X, Liu J, Wang T, Lu Q, Yin X. Mitochondria-dependent apoptosis was involved in the alleviation of Jujuboside A on diabetic kidney disease-associated renal tubular injury via YY1/PGC-1α signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156411. [PMID: 39884075 DOI: 10.1016/j.phymed.2025.156411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Renal tubular injury was a significant pathological change of diabetic kidney disease (DKD), and the amelioration of renal tubular injury through mitochondrial function was an important treatment strategy of DKD. Our previous study had revealed that Jujuboside A (Ju A), the main active substance isolated from Semen Ziziphi Spinosae (SZS), could restore renal function of diabetic mice. However, its protective mechanism against DKD remains unclear. PURPOSE To investigate the effects and the mechanism of Ju A against DKD-associated renal tubular injury. STUDY DESIGN AND METHODS The anti-apoptotic effect of Ju A and its protection effect on mitochondria dysfunction of renal tubular epithelial cells (RTECs) were examined in high glucose (HG)-cultured HK-2 cells, and in db/db mice. Subsequently, Network Pharmacology analysis, molecular docking, luciferase assay, chromatin immunoprecipitation (ChIP), Yin Yang 1 (YY1) overexpression lentiviral vector and peroxisome proliferator-activated receptor-γ coactlvator-1α (PGC-1α) specific agonist ZLN005 were all used to identify the protective mechanism of Ju A towards DKD-associated mitochondrial dysfunction of RTECs. RESULTS Ju A inhibited RTECs apoptosis and ameliorated mitochondria dysfunction of RTECs of diabetic mice, and HG-cultured HK-2 cells. YY1 was the potential target of Ju A against DKD-related mitochondrial dysfunction, and the down-regulation of YY1 induced by Ju A increased PGC-1α promoter activity, leading to the restored mitochondrial function of HG-treated HK-2 cells. Renal tubule specific overexpression of YY1 intercepted the renal protective effect of Ju A on diabetic mice via blocking PGC-1α-mediated restoration of mitochondrial function of RTECs. The in-depth mechanism research revealed that the protective effect of Ju A towards DKD-associated renal tubular injury was linked to the restored mitochondrial function through YY1/PGC-1α signaling, resulting in the inhibited apoptosis of RTECs in diabetic condition via inactivating CytC-mediated Caspase9/Caspase3 signaling. CONCLUSION Ju A through the inhibition of mitochondria-dependent apoptosis alleviated DKD-associated renal tubular injury via YY1/PGC-1α signaling.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Yuting Peng
- Department of Pharmacy, Xuzhou Oriental Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Yuting Shao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Dandan Pan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Qian Cheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China
| | - Sitong Qian
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Baojing Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Xia Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Junjie Liu
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Urology, The affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, PR China
| | - Tao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Pharmacy, The affiliated hospital of Xuzhou Medical University, Xuzhou 221006, PR China.
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China.
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, PR China.
| |
Collapse
|
16
|
Yu HC, Chung BH, Kim Y, Lee Y, Sim H, Lee S, Hwang HP, Yu HC, Jeon S, Maeng HJ, Shin D, Kang KP, Seo SY, Bae EJ, Park BH. p21-Activated Kinase 4 and Ischemic Acute Kidney Injury in Mice and Humans. J Am Soc Nephrol 2025:00001751-990000000-00576. [PMID: 40019790 DOI: 10.1681/asn.0000000649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/14/2025] [Indexed: 04/06/2025] Open
Abstract
Background AKI after ischemia-reperfusion remains a substantial perioperative challenge lacking effective treatment. p21-activated kinase 4 (PAK4), a downstream effector of Rho GTPase, has been explored in hepatic ischemia-reperfusion injury, but its role in renal ischemia-reperfusion is unknown. Methods Wild-type and proximal tubule–specific Pak4 knockout mice underwent 25 minutes of ischemia followed by 24 hours of reperfusion injury. Primary tubular cells and human kidney-2 cells were exposed to hypoxia-reoxygenation injury to investigate the in vitro effect of PAK4. Selective degradation of PAK4 was employed using proteolysis-targeting chimera (PROTAC) to ameliorate AKI. Results Post–ischemia-reperfusion, the expression of PAK4 was upregulated through hypoxia-inducible factor 1 α in mouse kidneys. Deletion of PAK4 in proximal tubule cells, but not in myeloid cells, significantly mitigated ischemia-reperfusion–induced AKI, as evidenced by decreased levels of BUN, creatinine, tubular necrosis, apoptosis, macrophage infiltration, and lipid accumulation compared with control mice. Further investigation revealed that PAK4 phosphorylated GSH peroxidase 3 (GPx3) at T47, leading to its proteasomal degradation. In addition, pretreatment of mice with the PAK4 PROTAC preserved GPx3 and enhanced fatty acid β-oxidation, thereby protecting against AKI. In kidney tissues from people with a kidney transplant, elevated levels of PAK4 protein and phosphorylation of GPx3 at T47 were observed. Conclusions Renal tubular PAK4 contributes to tissue damage during ischemia-reperfusion injury, whereas PAK4 PROTAC mitigates ischemia-reperfusion injury by reducing oxidative stress and promoting fatty acid β-oxidation.
Collapse
Affiliation(s)
- Hwang Chan Yu
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejon, Republic of Korea
| | - Byeoung Hoon Chung
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Yoejin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejon, Republic of Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyunchae Sim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hong Pil Hwang
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hee Chul Yu
- Department of Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seunggyu Jeon
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Han-Joo Maeng
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Eun Ju Bae
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, Republic of Korea
| | - Byung-Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejon, Republic of Korea
| |
Collapse
|
17
|
Lan SF, Yang ZH, Feng L, Wen YT, Chen KN, Fan LL, Wang MJ, Liu WT. MTDH inhibits CrAT to promote mitochondrial damage in palmitic acid-induced renal tubular cells. Acta Diabetol 2025:10.1007/s00592-025-02476-5. [PMID: 40100360 DOI: 10.1007/s00592-025-02476-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Mitochondrial dysfunction leading to impaired energy metabolism has been recognized as a pivotal factor contributing to renal tubular epithelial cells (RTECs) damage in the context of dyslipidemia conditions in diabetic kidney disease (DKD). The primary objective of this study is to elucidate the role and underlying mechanism of the proto-oncogene Metadherin (MTDH) in mediating mitochondrial damage within this specific pathological context in vitro. METHODS The expression of MTDH in RTECs was modulated by transfecting small interfering RNA and plasmid, while palmitic acid (PA) was employed to simulate diabetic lipid metabolism disorder. Mitochondrial damage was evaluated by examining various parameters including mitochondrial morphology, membrane potential, reactive oxygen species (ROS) production, adenosine triphosphate (ATP) production, as well as morphological and structural alterations. Additionally, Carnitine acetyltransferase (CrAT) expression was assessed using Western blotting and quantitative real-time polymerase chain reaction, and CrAT activity was quantified. RESULT MTDH expression was upregulated in PA-induced RTECs, while CrAT expression and activity were inhibited. Downregulation of MTDH mitigated PA-induced mitochondrial damage, as demonstrated by the preservation of mitochondrial membrane potential, reduction in mitochondrial ROS production, prevention of ATP depletion, and maintenance of mitochondrial structure. This was accompanied by an upregulation in CrAT expression and activity. Conversely, overexpression of MTDH exacerbated mitochondrial dysfunction by impairing membrane potential, augmenting mitochondrial ROS production, inhibiting ATP synthesis, and suppressing CrAT expression and activity. CONCLUSION In the context of dyslipidemia conditions, MTDH is upregulated and suppresses the expression and activity of CrAT in RTECs, thereby inducing mitochondrial dysfunction and perturbing energy metabolism. These alterations exacerbate the injury to RTECs, consequently promoting the progression of DKD.
Collapse
Affiliation(s)
- Shan-Fen Lan
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Zhen-Hua Yang
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Li Feng
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Yu-Ting Wen
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Kun-Ni Chen
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Lang-Lin Fan
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - Ming-Jun Wang
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China.
| | - Wen-Ting Liu
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China.
| |
Collapse
|
18
|
Yan L, Guo L. The role and mechanism of m6A methylation in diabetic nephropathy. Life Sci 2025; 363:123355. [PMID: 39778764 DOI: 10.1016/j.lfs.2024.123355] [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: 10/04/2024] [Revised: 12/19/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications of diabetes mellitus, characterized by progressive deterioration of renal structure and function, which may eventually lead to end-stage kidney disease (ESKD). The N6-methyladenosine (m6A) methylation, an important modality of RNA modification, involves three classes of key regulators, writers (e.g., METTL3), erasers (e.g., FTO, ALKBH5) and readers (e.g., YTHDF2), which play important roles in DN. Writers are responsible for introducing m6A modifications on RNAs, erasers remove m6A modifications and readers recognize and bind m6A-modified RNAs to regulate RNAs functions, such as mRNA stability, translation and localization. In DN, abnormal m6A modification may promote kidney injury and proteinuria by regulating key pathways involved in multiple processes, including lipid metabolism and inflammatory response, in kidney cells such as podocytes. Therefore, an in-depth study of the role and mechanism of m6A methylation that are regulated by "writers", "erasers" and "readers" in DN is expected to provide new targets and strategies for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Linjing Yan
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, PR China
| | - Liang Guo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, PR China.
| |
Collapse
|
19
|
Iheagwam FN, Joseph AJ, Adedoyin ED, Iheagwam OT, Ejoh SA. Mitochondrial Dysfunction in Diabetes: Shedding Light on a Widespread Oversight. PATHOPHYSIOLOGY 2025; 32:9. [PMID: 39982365 DOI: 10.3390/pathophysiology32010009] [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: 07/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 02/22/2025] Open
Abstract
Diabetes mellitus represents a complicated metabolic condition marked by ongoing hyperglycemia arising from impaired insulin secretion, inadequate insulin action, or a combination of both. Mitochondrial dysfunction has emerged as a significant contributor to the aetiology of diabetes, affecting various metabolic processes critical for glucose homeostasis. This review aims to elucidate the complex link between mitochondrial dysfunction and diabetes, covering the spectrum of diabetes types, the role of mitochondria in insulin resistance, highlighting pathophysiological mechanisms, mitochondrial DNA damage, and altered mitochondrial biogenesis and dynamics. Additionally, it discusses the clinical implications and complications of mitochondrial dysfunction in diabetes and its complications, diagnostic approaches for assessing mitochondrial function in diabetics, therapeutic strategies, future directions, and research opportunities.
Collapse
Affiliation(s)
- Franklyn Nonso Iheagwam
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amarachi Joy Joseph
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota 112104, Nigeria
| | - Eniola Deborah Adedoyin
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota 112104, Nigeria
| | | | - Samuel Akpoyowvare Ejoh
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota 112104, Nigeria
| |
Collapse
|
20
|
Zhao M, Cao Y, Ma L. New insights in the treatment of DKD: recent advances and future prospects. BMC Nephrol 2025; 26:72. [PMID: 39934650 PMCID: PMC11817338 DOI: 10.1186/s12882-025-03953-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
Diabetic kidney disease (DKD) represents the predominant and severe microvascular complication associated with diabetes, frequently culminating in End-Stage Kidney Disease (ESKD). The escalating prevalence of diabetes has correspondingly led to a rise in DKD incidence, imposing significant challenges on both individuals and society. The etiology of DKD is multifaceted and remains devoid of definitive therapeutic interventions. This article examines the pharmacological actions and mechanisms of different drugs used for the prevention and treatment of DKD that are currently in clinical use or undergoing development. The goal is to offer insights for early intervention based on therapeutic combinations to potentially slow kidney disease progression.
Collapse
Affiliation(s)
- Meimei Zhao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Yongtong Cao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China.
| | - Liang Ma
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
21
|
Zhu Z, Luan G, Wu S, Song Y, Shen S, Wu K, Qian S, Jia W, Yin J, Ren T, Ye J, Wei L. Single-cell atlas reveals multi-faced responses of losartan on tubular mitochondria in diabetic kidney disease. J Transl Med 2025; 23:90. [PMID: 39838394 PMCID: PMC11748887 DOI: 10.1186/s12967-025-06074-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Mitochondria are crucial to the function of renal tubular cells, and their dynamic perturbation in many aspects is an important mechanism of diabetic kidney disease (DKD). Single-nucleus RNA sequencing (snRNA-seq) technology is a high-throughput sequencing analysis technique for RNA at the level of a single cell nucleus. Here, our DKD mouse kidney single-cell RNA sequencing conveys a more comprehensive mitochondrial profile, which helps us further understand the therapeutic response of this unique organelle family to drugs. METHODS After high fat diet (HFD), mice were intraperitoneally injected with streptozotocin (STZ) to induce DKD, and then divided into three subsets: CON (healthy) subset, DKD (vehicle) subset, and LST (losartan; 25 mg/kg/day) subset. Divide HK-2 cell into LG (low glucose; 5 mM) and HG (high glucose; 30 mM) and HG + LST (losartan; 1 µ M) subsets. snRNA-seq was performed on the renal tissues of LST and DKD subset mice. To reveal the effects of losartan on gene function and pathway changes in renal tubular mitochondria, Gene Ontology (GO) enrichment analysis and GSEA/GSVA scoring were performed to analyze the specific response of proximal tubular (PT) cell mitochondria to losartan treatment, including key events in mitochondrial homeostasis such as mitochondrial morphology, dynamics, mitophagy, autophagic flux, mitochondrial respiratory chain, apoptosis, and ROS generation. Preliminary validation through in vitro and in vivo experiments, including observation of changes in mitochondrial morphology and dynamics using probes such as Mitotracker Red, and evaluation of the effect of losartan on key events of mitochondrial homeostasis perturbation using electron microscopy, laser confocal microscopy, immunofluorescence, and Western blotting. Detection of autophagic flux in cells by transfecting Ad-mCherry-GFP-LC3B dual fluorescence labeled adenovirus. Various fluorescent probes and energy detector are used to detect mitochondrial apoptosis, ROS, and respiration of mitochondrion. RESULTS Through the single-cell atlas of DKD mouse kidneys, it was found that losartan treatment significantly increased the percentage of PT cells. Gene Ontology (GO) enrichment analysis of differentially expressed genes showed enrichment of autophagy of mitochondrion pathway. Further GSEA analysis and GSVA scoring revealed that mitophagy and other key mitochondrial perturbation events, such as ROS production, apoptosis, membrane potential, adenosine triphosphate (ATP) synthesis, and mitochondrial dynamics, were involved in the protective mechanism of losartan on PT cells, thereby improving mitochondrial homeostasis. Consistent results were also obtained in mice and cellular experiments. In addition, we highlighted a specific renal tubular subpopulation with mitophagy phenotype found in single-cell data, and preliminarily validated it with co-localization and increased expression of Pink1 and Gclc in kidney specimens of DKD patients treated with losartan. CONCLUSIONS Our research suggests that scRNA-seq can reflect the multifaceted mitochondrial landscape of DKD renal tubular cells after drug treatment, and these findings may provide new targets for DKD therapy at the organelle level.
Collapse
Affiliation(s)
- Zhen Zhu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Guangxin Luan
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Song Wu
- Department of Cardiothoracic Surgery, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Yiyi Song
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Shuang Shen
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Kaiyue Wu
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Shengnan Qian
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China
| | - Jun Yin
- Department of Endocrine Medicine, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, Chin, China.
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China.
| | - Jianping Ye
- Shanghai Diabetes Institute, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, China.
| | - Li Wei
- Department of Endocrine Medicine, Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201306, Chin, China.
| |
Collapse
|
22
|
Xue M, Tian Y, Zhang H, Dai S, Wu Y, Jin J, Chen J. Curcumin nanocrystals ameliorate ferroptosis of diabetic nephropathy through glutathione peroxidase 4. Front Pharmacol 2025; 15:1508312. [PMID: 39834811 PMCID: PMC11743454 DOI: 10.3389/fphar.2024.1508312] [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: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Objective The aim of this study was to investigate the effect of curcumin nanocrystals (Cur-NCs) on ferroptosis in high-glucose (HG)-induced HK-2 cells and streptozotocin (STZ)-induced diabetic nephropathy model (DN) rats. The purpose is to determine whether Cur NCs can become a promising treatment option for diabetes nephropathy by reducing ferroptosis. Methods Cur-NCs were prepared using microfluidic technology and studied using dynamic light scattering and transmission electron microscopy. HK-2 cells were treated with 30 mM HG to create a renal tubule damage cell model. Then, cell viability was evaluated in HK-2 cells treated with varying concentrations of Cur-NCs (0.23, 0.47, 0.94, 1.87, 3.75, 7.5, 15, and 30 μg/mL) using Cell Counting Kit-8 (CCK-8). Furthermore, in vivo experiments were carried out to investigate the roles of Cur-NCs in STZ-induced DN rats. Results The results showed that HG treatment greatly enhanced the levels of LDH, MDA, Iron, lipid ROS, apoptosis, NCOA4, TFR-1, while decreasing the expression of GSH, GPX4, SLC7A11, and FTH-1. These effects induced by HG could be attenuated by Cur-NCs. Cur-NCs also reduced the HG-induced decrease in cell viability, as well as the increase in lipid ROS and cell apoptosis, however erastin could inhibit their effects. Furthermore, the in vivo results showed that Cur-NCs reduced ferroptosis and inhibited renal damage in DN rats. Conclusion This study demonstrates that Cur-NCs can significantly attenuate ferroptosis in a STZ-induced renal damage model by recovering GPX4, implying that Cur-NCs may be a promising therapy option for DN.
Collapse
Affiliation(s)
- Mengjiao Xue
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yiwei Tian
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, China
| | - Hua Zhang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Shijie Dai
- College of Life Science, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangsheng Wu
- College of Life Science, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Juan Jin
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jian Chen
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
23
|
Yan Q, Du Y, Huang F, Zhang Q, Zhan M, Wu J, Yan J, Zhang P, Lin H, Han L, Huang X. Identification of mitochondria-related genes as diagnostic biomarkers for diabetic nephropathy and their correlation with immune infiltration: New insights from bioinformatics analysis. Int Immunopharmacol 2024; 142:113114. [PMID: 39265357 DOI: 10.1016/j.intimp.2024.113114] [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: 06/29/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a common and severe microvascular complication of diabetes. Mitochondrial dysfunction and immune inflammation are important factors in the pathogenesis of DN. However, the specific mechanisms and their intricate interactions in DN remain unclear. Besides, there are no effective specific predictive or diagnostic biomarkers for DN so far. Therefore, this study aims to elucidate the role of mitochondrial-related genes and their possibility as predictive or diagnostic biomarkers, as well as their crosstalk with immune infiltration in the progression of DN. METHODS Based on the GEO database and limma R package, the differentially expressed genes (DEGs) of DN were identified. Mitochondrial-related DEGs (MitoDEGs) were then obtained by intersecting these DEGs with mitochondria-related genes from the MitoCarta 3.0 database. Subsequently, the candidate hub genes were further screened by gene co-expression network analysis (WGCNA), and verified mRNA levels of these genes by real-time quantitative PCR (qRT-PCR) in high-glucose-treated human proximal tubular (HK-2) cells. The verified hub genes were utilized to construct a combined diagnostic model for DN, with its diagnostic efficacy assessed across the GSE30122 and GSE96804 datasets. Additionally, the immune infiltration pattern in DN was assessed with the CIBERSORT algorithm, and the Nephroseq v5 database was used to analyze the correlation between hub genes and clinical features of DN. RESULTS Seven mitochondria-related candidate hub genes were screened from 56 MitoDEGs. Subsequently, the expression levels of six of them, namely EFHD1, CASP3, AASS, MPC1, NT5DC2, and BCL2A1, exhibited significant inter-group differences in the HK-2 cell model. The diagnostic model based on the six genes demonstrated good diagnostic efficacy in both training and validation sets. Furthermore, correlation analysis indicated that EFHD1 and AASS, downregulated in DN, are positively correlated with eGFR and negatively with serum creatinine. Conversely, CASP3, NT5DC2, and BCL2A1, upregulated in DN, show opposite correlations. In addition, spearman analysis revealed that the six hub genes were significantly associated with the infiltration of immune cells, including M1 and M2 macrophages, mast cells, resting NK cells, gamma delta T cells, and follicular helper T cells. CONCLUSION This study elucidated the characteristics of mitochondria-related genes and their correlation with immune cell infiltration in DN, providing new insights for exploring the pathogenesis of DN and facilitating the identification of new potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Qiaofang Yan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuanyuan Du
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Fei Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qiaoxuan Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Min Zhan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Junbiao Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Jun Yan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Pengwei Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, 510120, China
| | - Haibiao Lin
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Liqiao Han
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China.
| | - Xianzhang Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China.
| |
Collapse
|
24
|
Barutta F, Corbetta B, Bellini S, Gambino R, Bruno S, Kimura S, Hase K, Ohno H, Gruden G. Protective effect of mesenchymal stromal cells in diabetic nephropathy: the In vitro and In vivo role of the M-Sec-tunneling nanotubes. Clin Sci (Lond) 2024; 138:1537-1559. [PMID: 39535903 DOI: 10.1042/cs20242064] [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: 09/19/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/16/2024]
Abstract
Mitochondrial dysfunction plays an important role in the development of podocyte injury in diabetic nephropathy (DN). Tunnelling nanotubes (TNTs) are long channels that connect cells and allow organelle exchange. Mesenchymal stromal cells (MSCs) can transfer mitochondria to other cells through the M-Sec-TNTs system. However, it remains unexplored whether MSCs can form heterotypic TNTs with podocytes, thereby enabling the replacement of diabetes-damaged mitochondria. In this study, we analysed TNT formation, mitochondrial transfer, and markers of cell injury in podocytes that were pre-exposed to diabetes-related insults and then co-cultured with diabetic or non-diabetic MSCs. Furthermore, to assess the in vivo relevance, we treated DN mice with exogenous MSCs, either expressing or lacking M-Sec, carrying fluorescent-tagged mitochondria. MSCs formed heterotypic TNTs with podocytes, allowing mitochondrial transfer, via a M-Sec-dependent mechanism. This ameliorated mitochondrial function, nephrin expression, and reduced apoptosis in recipient podocytes. However, MSCs isolated from diabetic mice failed to confer cytoprotection due to Miro-1 down-regulation. In experimental DN, treatment with exogenous MSCs significantly improved DN, but no benefit was observed in mice treated with MSCs lacking M-Sec. Mitochondrial transfer from exogenous MSCs to podocytes occurred in vivo in a M-Sec-dependent manner. These findings demonstrate that the M-Sec-TNT-mediated transfer of mitochondria from healthy MSCs to diabetes-injured podocytes can ameliorate podocyte damage. Moreover, M-Sec expression in exogenous MSCs is essential for providing renoprotection in vivo in experimental DN.
Collapse
Affiliation(s)
- Federica Barutta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Stefania Bellini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Shunsuke Kimura
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
25
|
Shao M, Chen J, Zhang F, Su Q, Lin X, Wang W, Chen C, Ren H, Zheng S, Hui S, Qin S, Ni Y, Zhong J, Yang J. 4-Octyl itaconate attenuates renal tubular injury in db/db mice by activating Nrf2 and promoting PGC-1α-mediated mitochondrial biogenesis. Ren Fail 2024; 46:2403653. [PMID: 39291665 PMCID: PMC11411562 DOI: 10.1080/0886022x.2024.2403653] [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: 01/02/2024] [Revised: 07/11/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024] Open
Abstract
Objectives: The aim of this study was to investigate the mechanism of itaconate's potential effect in diabetic kidney disease. Methods: Renal immune responsive gene 1 (IRG1) levels were measured in db/db mice and streptozotocin (STZ) + high-fat diet (HFD)-induced diabetic mice. Irg1 knockout mice were generated. db/db mice were treated with 4-octyl itaconate (4-OI, 50 mg/kg), a derivative of itaconate, for 4 weeks. Renal function and morphological changes were investigated. Ultrastructural alterations were determined by transmission electron microscopy. Results: Renal IRG1 levels were reduced in two diabetic models. STZ+HFD-treated Irg1 knockout mice exhibited aggravated renal tubular injury and worsened renal function. Treatment with 4-OI lowered urinary albumin-to-creatinine ratio and blood urea nitrogen levels, and restored renal histological changes in db/db mice. It improved mitochondrial damage, increased expressions of peroxisome-proliferator-activated receptor γ coactivator-1α (PGC-1α) and mitochondrial transcription factor A (TFAM) in the renal cortex of db/db mice. These were confirmed in vitro; 4-OI improved high glucose-induced abnormal mitochondrial morphology and TFAM expression in HK-2 cells, effects that were inhibited by PGC-1α silencing. Moreover, 4-OI reduced the number of apoptotic cells in the renal cortex of db/db mice. Further study showed that 4-OI increased renal Nrf2 expression and decreased oxidative stress levels in db/db mice. In HK-2 cells, 4-OI decreased high glucose-induced mitochondrial ROS production, which was reversed by Nrf2 silencing. Nrf2 depletion also inhibited 4-OI-mediated regulation of PGC-1α, TFAM, and mitochondrial apoptotic protein expressions. Conclusions: 4-OI attenuates renal tubular injury in db/db mice by activating Nrf2 and promoting PGC-1α-mediated mitochondrial biogenesis.
Collapse
Affiliation(s)
- Muqing Shao
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiayao Chen
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fuwei Zhang
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Su
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqian Lin
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiwei Wang
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Suocheng Hui
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si Qin
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yinxing Ni
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Zhong
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Yang
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Hu H, Hu J, Chen Z, Yang K, Zhu Z, Hao Y, Zhang Z, Li W, Peng Z, Cao Y, Sun X, Zhang F, Chi Q, Ding G, Liang W. RBBP6-Mediated ERRα Degradation Contributes to Mitochondrial Injury in Renal Tubular Cells in Diabetic Kidney Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405153. [PMID: 39441040 PMCID: PMC11633482 DOI: 10.1002/advs.202405153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Diabetic Kidney Disease (DKD), a major precursor to end-stage renal disease, involves mitochondrial dysfunction in proximal renal tubular cells (PTCs), contributing to its pathogenesis. Estrogen-related receptor α (ERRα) is essential for mitochondrial integrity in PTCs, yet its regulation in DKD is poorly understood. This study investigates ERRα expression and its regulatory mechanisms in DKD, assessing its therapeutic potential. Using genetic, biochemical, and cellular approaches, ERRα expression Was examined in human DKD specimens and DKD mouse models. We identified the E3 ubiquitin ligase retinoblastoma binding protein 6 (RBBP6) as a regulator of ERRα, promoting its degradation through K48-linked polyubiquitination at the K100 residue. This degradation pathway significantly contributed to mitochondrial injury in PTCs of DKD models. Notably, conditional ERRα overexpression or RBBP6 inhibition markedly reduced mitochondrial damage in diabetic mice, highlighting ERRα's protective role in maintaining mitochondrial integrity. The interaction between RBBP6 and ERRα opens new therapeutic avenues, suggesting that modulating RBBP6-ERRα interactions could be a strategy for preserving mitochondrial function and slowing DKD progression.
Collapse
Affiliation(s)
- Hongtu Hu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Jijia Hu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Zhaowei Chen
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Keju Yang
- The First College of Clinical Medical ScienceChina Three Gorges UniversityYichang443000China
| | - Zijing Zhu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Yiqun Hao
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Zongwei Zhang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Weiwei Li
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Zhuan Peng
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Yun Cao
- Department of NephrologyHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical College)Haikou100053China
| | - Xiaoling Sun
- Ultrastructural Pathology CenterRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Fangcheng Zhang
- Ultrastructural Pathology CenterRenmin Hospital of Wuhan UniversityWuhan430060China
| | - Qingjia Chi
- Department of Mechanics and Engineering StructureWuhan University of TechnologyWuhan430070China
| | - Guohua Ding
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| | - Wei Liang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Key Clinical Research Center of Kidney DiseaseWuhan430060China
| |
Collapse
|
27
|
Huang Q, Zhong K, Wei J. CircPWWP2A promotes renal interstitial fibrosis through modulating miR-182/ROCK1 axis. Ren Fail 2024; 46:2396455. [PMID: 39229866 PMCID: PMC11376294 DOI: 10.1080/0886022x.2024.2396455] [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: 01/02/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
Renal fibrosis is a long-term and progressively worsening condition that impacts kidney function during aging and in the context of chronic kidney disease (CKD). CKD and renal fibrosis affect approximately 10% of the global population and are prevalent in about half of individuals over the age of 70. Despite ongoing research, the mechanisms underlying renal fibrosis are still not well understood, and there is currently a lack of effective treatments available. In the present study, we demonstrated a significant increase of circPWWP2A in renal tubular cells both in vivo and in vitro models of renal fibrosis. Suppressing circPWWP2A has the potential to reduce mitochondrial dysfunction and the production of mitochondrial reactive oxygen species (mtROS), ultimately leading to the inhibition of renal fibrosis. Whereas, supplementation of circPWWP2A led to more serve mitochondrial dysfunction, mtROS production and renal fibrosis. Mechanistically, we found the expression of circPWWP2A was negatively correlated with the expression of miR-182. And we further confirmed miR-182 was the direct target of circPWWP2A by dual-luciferase reporter assay and RIP assay. Then, we found miR-182 suppressed the expression of ROCK1 in both in vitro and in vivo models of renal fibrosis. Luciferase microRNA target reporter assay further indicated ROCK1 as a direct target of miR-182. Knockdown of ROCK1 inhibits renal fibrosis and mitochondrial dysfunction, suggesting ROCK1 not only served as an injurious role in mitochondrial homeostasis but also a pro-fibrotic factor in CKD. Taking together, our findings suggest that circPWWP2A may promote renal interstitial fibrosis by modulating miR-182/ROCK1-mediated mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qian Huang
- Department of Nephrology, Haikou Third People’s Hospital, Haikou, Hainan, China
| | - Kaiyi Zhong
- Department of Nephrology, Danzhou West Central Hospital, Danzhou, Hainan, China
| | - Jiali Wei
- Department of Nephrology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical College), Haikou, China
| |
Collapse
|
28
|
Zhu Y, Yang B, Chen S, Chen G, Zeng X, Min H, Xu L. M6A RNA Methylation-Mediated TUG1 Stability Maintains Mitochondrial Homeostasis during Kidney Aging by Epigenetically Regulating PGC1-α Expression. Antioxid Redox Signal 2024; 41:993-1013. [PMID: 39135383 DOI: 10.1089/ars.2024.0631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Background: Aging is a significant risk factor for the increased incidence of acute kidney injury and chronic kidney disease, posing significant challenges to global public health. The role of N6-methyladenosine (m6A) in the development of chronic kidney disease has been reported, but the regulatory mechanism of m6A in kidney aging remains unclear. Results: In this study, we identified a long noncoding RNA (lncRNA), called taurine up-regulated 1 (TUG1), which exhibited a significantly decreased level of m6A modification in human aged kidney through the m6A-lncRNA epitranscriptome microarray. Bioinformatics analysis and machine learning predicted that TUG1 had potentially strong interaction with PGC1-α. RNA immunoprecipitation and chromatin immunoprecipitation analysis showed that TUG1 promoted proliferator-activated receptor γ coactivator-1α (PGC1-α) expression by directly interacting with its TUG-1 binding element region, thereby impacting mitochondrial quality control (MQC), cellular senescence, and renal fibrosis. Silencing the RNA m6A methylase methyltransferase 14 (METTL14) or the reader protein insulin-like growth factor 2 mRNA-binding proteins (IGF2BP2) resulted in the weakened stability of lncRNA TUG1, contributing to an imbalance in MQC. Conclusion: Our study demonstrated that the m6A modification and stability of TUG1 were mediated by METTL14 in an IGF2BP2-dependent manner, and modulate the mitochondrial homeostasis in kidney aging by direct targeting PGC-1α. These findings provide a new perspective on potential therapeutic targets for kidney aging. Antioxid. Redox Signal. 41, 993-1013.
Collapse
Affiliation(s)
- Yonghong Zhu
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bowen Yang
- Department of Medical Oncology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Suyun Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guanqing Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaobian Zeng
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Li Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
29
|
Huang R, Zhang C, Xiang Z, Lin T, Ling J, Hu H. Role of mitochondria in renal ischemia-reperfusion injury. FEBS J 2024; 291:5365-5378. [PMID: 38567754 DOI: 10.1111/febs.17130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/30/2024] [Accepted: 03/22/2024] [Indexed: 12/19/2024]
Abstract
Acute kidney injury (AKI) induced by renal ischemia-reperfusion injury (IRI) has a high morbidity and mortality, representing a worldwide problem. The kidney is an essential organ of metabolism that has high blood perfusion and is the second most mitochondria-rich organ after the heart because of the high ATP demands of its essential functions of nutrient reabsorption, acid-base and electrolyte balance, and hemodynamics. Thus, these energy-intensive cells are particularly vulnerable to mitochondrial dysfunction. As the bulk of glomerular ultrafiltrate reabsorption by proximal tubules occurs via active transport, the mitochondria of proximal tubules must be equipped for detecting and responding to fluctuations in energy availability to guarantee efficient basal metabolism. Any insults to mitochondrial quality control mechanisms may lead to biological disruption, blocking the clearance of damaged mitochondria and resulting in morphological change and tissue dysfunction. Extensive research has shown that mitochondria have pivotal roles in acute kidney disease, so in this article, we discuss the role of mitochondria, their dynamics and mitophagy in renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Zhengjie Xiang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Tao Lin
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Jian Ling
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, China
| |
Collapse
|
30
|
Zhu Y, Dong C, Xu Z, Lou Y, Tian N, Guan Y, Nie P, Luo M, Luo P. Human Umbilical Cord Mesenchymal Stem Cells Alleviate Diabetic Nephropathy by Inhibiting Ferroptosis via the JNK/KEAP1/NRF2 Signaling Pathway. Antioxid Redox Signal 2024. [PMID: 39602247 DOI: 10.1089/ars.2024.0575] [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] [Indexed: 11/29/2024]
Abstract
Aims: Diabetic nephropathy (DN) is a major cause of end-stage renal disease, with no therapeutic interventions available to control its progression. Ferroptosis, an iron-dependent regulated cell death characterized by lipid peroxidation, plays a pivotal role in the pathogenesis of DN. Human umbilical cord mesenchymal stem cells (hUCMSCs) are an effective treatment modality for DN; however, the underlying mechanism of action remains unclear. The aim of the present study was to investigate whether hUCMSCs alleviate DN via inhibiting ferroptosis and its molecular mechanisms in type 2 diabetic mice and high-glucose and palmitate-stimulated human renal tubular epithelial cell (HK-11) models. Results: Our findings revealed that hUCMSCs improved the renal structure and function and tubular injuries. HUCMSC treatment can inhibit ferroptosis by decreasing iron content, reducing reactive oxygen species, malondialdehyde and 4-hydroxynonenal generation, decreasing the expression of positive ferroptosis mediator transferrin receptor 1 and long-chain acyl-CoA synthetase 4, and enhancing the expression of negative ferroptosis mediators (i.e., ferritin heavy chain, glutathione peroxidase 4, and system Xc-cystine/glutamate reverse transporter). Mechanistically, hUCMSC treatment inhibited c-Jun N-terminal kinase (JNK) and Kelch-like ECH-associated protein 1 (KEAP1) activation while increasing the expression of nuclear factor erythroid 2-related factor 2 (NRF2). Furthermore, pretreatment of HK-11 cells with NRF2 siRNA, the JNK inhibitor SP600125, or the JNK agonist anisomycin demonstrated the regulation of the JNK/KEAP1/NRF2 signaling pathway by hUCMSCs. Innovation and Conclusion: HUCMSCs inhibit ferroptosis in DN via the JNK/KEAP1/NRF2 signaling pathway, providing a new perspective and scientific evidence for treating DN. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Changqing Dong
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Zhiheng Xu
- Department of Radiology, Changchun Stomatological Hospital, Changchun, P.R. China
| | - Yan Lou
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Na Tian
- Research and Development Department, Jilin Tuohua Biotechnology Co., Ltd., Siping, P.R. China
| | - Yucan Guan
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
31
|
Yu S, Lu X, Li C, Han Z, Li Y, Zhang X, Guo D. TFAM and Mitochondrial Protection in Diabetic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:4355-4365. [PMID: 39588133 PMCID: PMC11586499 DOI: 10.2147/dmso.s487815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic kidney disease (DKD) is a significant complication of diabetes and a major cause of end-stage renal disease. Affecting around 40% of diabetic patients, DKD poses substantial economic burdens due to its prevalence worldwide. The primary clinical features of DKD include the leakage of proteins into the urine, altered glomerular filtration, and an increased risk of cardiovascular diseases. Current treatments focus on managing hypertension and hyperglycemia to slow the progression of DKD. These include the use of SGLT2 inhibitors to control blood sugar and ACE inhibitors to reduce blood pressure. Despite these measures, current treatments do not cure DKD and fail to address its underlying causes. Emerging research highlights mitochondrial dysfunction as a pivotal factor in DKD progression. The kidneys' high energy requirements make them particularly susceptible to disturbances in mitochondrial function. In DKD, mitochondrial damage leads to reduced energy production and increased oxidative stress, exacerbating tissue damage. Mitochondrial DNA (mtDNA) damage is a key aspect of this dysfunction, with studies suggesting that changes in mtDNA copy number can serve as biomarkers for the progression of the disease. Efforts to target mitochondrial dysfunction are gaining traction as a potential therapeutic strategy. This includes promoting mitochondrial health through pharmacological and lifestyle interventions aimed at enhancing mitochondrial function and reducing oxidative stress. Such approaches could lead to more effective treatments that directly address the DKD.
Collapse
Affiliation(s)
- Siming Yu
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, People’s Republic of China
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xinxin Lu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chunsheng Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zehui Han
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yue Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xianlong Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Dandan Guo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
32
|
Wang Y, Yu L, Li Y, Cha S, Shi L, Wang J, Ge F, Huang C, Huang H, Tu Y, Wan Y, Shen S. Supplemented Gegen Qinlian Decoction Formula attenuates podocyte mitochondrial fission and renal fibrosis in diabetic kidney disease by inhibiting TNF-α-mediated necroptosis, compared with empagliflozin. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118572. [PMID: 39025164 DOI: 10.1016/j.jep.2024.118572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Recently, podocyte mitochondrial dysfunction and necroptosis have been shown to play critical roles in renal fibrosis (RF) in diabetic kidney disease (DKD); however, these conditions lack effective treatment. In China, the supplemented Gegen Qinlian Decoction Formula (SGQDF), which originates from the classical prescription Gegen Qinlian Decoction, has been widely used to treat patients with DKD. However, it remains unclear whether SGQDF alleviates podocyte injury-associated RF in patients with DKD. AIM OF STUDY This study aimed to clarify the therapeutic effects of SGQDF compared with those of empagliflozin (EMPA) on podocyte mitochondrial fission and RF in DKD and its necroptosis-related mechanisms. MATERIALS AND METHODS Modified DKD rat models were developed through a combination of uninephrectomy, streptozotocin administration through intraperitoneal injection, and exposure to a high-fat diet. Following RF formation, the DKD rat models received either a high dose of SGQDF (H-SGQDF), a low dose of SGQDF (L-SGQDF), EMPA, or vehicle for 4 weeks. In our in vitro study, we subjected cultured murine podocytes to a high-glucose environment and various treatments including Mdivi-1, adalimumab, and necrostatin-1, with or without H-SGQDF or EMPA. SGQDF target prediction and molecular docking verification were performed. For the in vivo study, we focused on examining changes in the parameters associated with renal injury, RF, and oxidative stress (OS)-induced injuries in podocytes. Both in vivo and in vitro studies included an analysis of changes in podocyte mitochondrial fission, TNF-α-induced podocyte necroptosis, and the RIPK1/RIPK3/MLKL signaling pathway activation. RESULTS SGQDF improved renal injury markers, including body weight, blood glucose, serum creatinine, blood urea nitrogen, and urinary albumin, in a dose-dependent manner. The beneficial effects of H-SGQDF in vivo were greater than those of L-SGQDF alone in vivo. Interestingly, similar to EMPA, H-SGQDF ameliorated RF and reduced OS-induced podocyte injury in diabetic kidneys. Furthermore, TNF-α signaling was shown to be important in the network construction of "the SGQDF-component-target." Based on this, we also showed that the beneficial effects in vivo and in vitro of H-SGQDF were closely related to the improvement in mitochondrial dysfunction and the inhibition of TNF-α-induced necroptosis in podocytes. CONCLUSION In the present study, we showed that H-SGQDF, similar to EMPA, attenuates podocyte mitochondrial fission and RF, and that the underlying therapeutic mechanisms are closely related to inhibiting the activation of the RIPK1/RIPK3/MLKL signaling axis in diabetic kidneys. Our findings provide new pharmacological evidence for the application of H-SGQDF in the RF treatment of DKD.
Collapse
Affiliation(s)
- Yu Wang
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China; Institute of Chinese Medicine, Nanjing University, Nanjing, 210008, China; Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Lu Yu
- Department of Endocrinology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Yajing Li
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Siyu Cha
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Lijing Shi
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinlong Wang
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Fengfeng Ge
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Chubo Huang
- International Jingfang Institute, Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Huang Huang
- International Jingfang Institute, Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yue Tu
- Department of Traditional Chinese Medicine Health Preservation, Acupuncture, Moxibustion and Massage College, Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yigang Wan
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China; Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Shanmei Shen
- Department of Endocrinology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China; Department of Endocrinology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
33
|
Hou T, Jiang Y, Zhang J, Hu R, Li S, Fan W, Chen R, Zhang L, Li R, Qin L, Gu W, Wu Y, Zhang L, Zeng X, Sun Q, Mao Y, Liu C. Kidney Injury Evoked by Fine Particulate Matter: Risk Factor, Causation, Mechanism and Intervention Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403222. [PMID: 39316383 DOI: 10.1002/advs.202403222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Fine particulate matter (PM2.5) is suggested to pose a severe risk to the kidneys by inducing functional degradation and chronic kidney diseases (CKD). This study aims to explore the nephrotoxicity of PM2.5 exposure and the underlying mechanism. Herein, based on the UK Biobank, it is found that per interquartile range (IQR) increase in PM2.5 is associated with a 6% (95% CI: 1%-11%), 7% (95% CI: 3%-11%), 9% (95% CI: 4%-13%), 11% (95% CI: 9%-13%), and 10% (95% CI: 8%-12%) increase in the risk of nephritis, hydronephrosis, kidney stone, acute renal failure, and CKD, respectively. In experimental study, noticeable kidney injury, which is the initiation of kidney diseases, is observed with PM2.5 exposure in C57BL/6N mice (n = 8), accompanied with oxidative stress, autophagy and pyroptosis. In vitro, HK-2 cells with PM2.5-stimulation exhibit tubulopathy, increased reactive oxygen species (ROS) generation and activated pyroptosis and autophagy. All changes are abolished by ROS scavenger of N-acetyl-L-cysteine (NAC) both in vivo and in vitro. In conclusion, the study provides evidence showing that PM2.5 exposure is associated with 5 kinds of kidney diseases by directly inducing nephrotoxicity, in which ROS may be the potential target by triggering autophagy and pyroptosis.
Collapse
Affiliation(s)
- Tong Hou
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Yuqing Jiang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiyang Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Sanduo Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Li Qin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Yue Wu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Lina Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Xiang Zeng
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Yingying Mao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| |
Collapse
|
34
|
Liu CC, Ji JL, Wang Z, Zhang XJ, Ding L, Zhang Y, Zhou Y, Zhang DJ, Tang ZL, Cao JY, Zhang AQ, Liu BC, Li ZL, Ma RX. TRPC6-Calpain-1 Axis Promotes Tubulointerstitial Inflammation by Inhibiting Mitophagy in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:3301-3317. [PMID: 39534194 PMCID: PMC11551102 DOI: 10.1016/j.ekir.2024.08.019] [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: 04/20/2024] [Revised: 07/23/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Renal tubulointerstitial inflammation represents an effective indicator for predicting the progression of diabetic kidney disease (DKD). Mitophagy abnormality is 1 of the most important factors involved in tubule injury. However, the exact molecular mechanism underlying mitophagy abnormality-mediated tubulointerstitial inflammation in DKD remains poorly understood. Methods In this study, a streptozotocin-induced DKD mouse model was established and HK-2 cells treated with high glucose (HG) served as an in vitro model. Tubular mitophagy was regulated through pharmacological urolithin A (UA) administration. The functional effect of the transient receptor potential cation channel, subfamily C, member 6 (TRPC6) was explored using genetic interventions in vivo and in vitro. Results We found that renal tubulointerstitial inflammation in DKD was closely associated with mitophagy inhibition, which was mediated by disturbance of PINK1/Parkin pathway. Mitophagy activation significantly attenuated tubular injury and tubulointerstitial inflammation. Further, it was found that TRPC6 was markedly increased in DKD and played an essential role in mitophagy inhibition by activating calpain-1. Knockdown of Trpc6 partially reversed mitophagy abnormality and consequently attenuated tubular injury and tubulointerstitial inflammation in vivo and in vitro. Finally, we found that tubular TRPC6-mediated mitophagy inhibition was blocked with BAPTA (a specific Ca2+ chelator) or calpeptin (a specific calpain-1 inhibitor). Conclusion Our study reveals that TRPC6-calpain-1 axis promotes tubulointerstitial inflammation in DKD by inhibiting mitophagy.
Collapse
Affiliation(s)
- Cong-Cong Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jia-Ling Ji
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ze Wang
- Department of Nephrology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xing-Jian Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lin Ding
- Department of Nephrology, Minda Hospital Affiliated to Hubei Minzu University, Enshi, Hubei, China
| | - Yao Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Zhou
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Dong-Jie Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhen-Lin Tang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jing-Yuan Cao
- Department of Nephrology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ai-Qing Zhang
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Rui-Xia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
35
|
Sun L, Shao Y, Zhuang Z, Liu Z, Liu M, Qu C, Yang H. Targeting TGR5 to mitigate liver fibrosis: Inhibition of hepatic stellate cell activation through modulation of mitochondrial fission. Int Immunopharmacol 2024; 140:112831. [PMID: 39111149 DOI: 10.1016/j.intimp.2024.112831] [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: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024]
Abstract
Chronic hepatitis B virus (HBV) infection continues to be a prominent cause of liver fibrosis and end-stage liver disease in China, necessitating the development of effective therapeutic strategies. This study investigated the potential of targeting TGR5 to alleviate liver fibrosis by impeding the activation of hepatic stellate cells (HSCs), which play a pivotal role in fibrotic progression. Using the human hepatic stellate cell line LX-2 overexpressing hepatitis B virus X protein (HBX), this study revealed that TGR5 activation through INT-777 inhibits HBX-induced LX-2 cell activation, thereby ameliorating liver fibrosis, which is associated with the attenuation of mitochondrial fission and introduces a novel regulatory pathway in liver fibrosis. Additional experiments with mitochondrial fission inducers and inhibitors confirm the crucial role of mitochondrial dynamics in TGR5-mediated effects. In vivo studies using TGR5 knockout mice substantiate these findings, demonstrating exacerbated fibrosis in the absence of TGR5 and its alleviation with the mitochondrial fission inhibitor Mdivi-1. Overall, this study provides insights into TGR5-mediated regulation of liver fibrosis through the modulation of mitochondrial fission in HSCs, suggesting potential therapeutic strategies for liver fibrosis intervention.
Collapse
Affiliation(s)
- Li Sun
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China
| | - Yuancheng Shao
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China
| | - Zehao Zhuang
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China; Department of General Surgery, Second People's Hospital, Jintan District, Changzhou City, Jiangsu Province 213100, China
| | - Zhixin Liu
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China
| | - Mingjun Liu
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China; Department of Graduate School, Dalian Medical University, Dalian City, Liaoning Province 116011, China
| | - Chang Qu
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China
| | - Haojun Yang
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province 213100, China.
| |
Collapse
|
36
|
Li H, Xia Y, Zha H, Zhang Y, Shi L, Wang J, Huang H, Yue R, Hu B, Zhu J, Song Z. Dapagliflozin attenuates AKI to CKD transition in diabetes by activating SIRT3/PGC1-α signaling and alleviating aberrant metabolic reprogramming. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167433. [PMID: 39067538 DOI: 10.1016/j.bbadis.2024.167433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Patients with diabetes are prone to acute kidney injury (AKI) with a high mortality rate, poor prognosis, and a higher risk of progression to chronic kidney disease than non-diabetic patients. METHODS Streptozotocin (STZ)-treated type 1 and db/db type 2 diabetes model were established, AKI model was induced in mice by ischemia-reperfusion injury(IRI). Mouse proximal tubular cell cells were subjected to high glucose and hypoxia-reoxygenation in vitro. Transcriptional RNA sequencing was performed for clustering analysis and target gene screening. Renal structural damage was determined by histological staining, whereas creatinine and urea nitrogen levels were used to measure renal function. RESULTS Deteriorated renal function and renal tissue damage were observed in AKI mice with diabetic background. RNA sequencing showed a decrease in fatty acid oxidation (FAO) pathway and an increase in abnormal glycolysis. Treatment with Dapa, Sitagliptin(a DPP-4 inhibitor)and insulin reduced blood glucose levels in mice, and improved renal function. However, Dapa had a superior therapeutic effect and alleviated aberrant FAO and glycosis. Dapa reduced cellular death in cultured cells under high glucose hypoxia-reoxygenation conditions, alleviated FAO dysfunction, and reduced abnormal glycolysis. RNA sequencing showed that SIRT3 expression was reduced in diabetic IRI, which was largely restored by Dapa intervention. 3-TYP, a SIRT3 inhibitor, reversed the renal protective effects of Dapa and mediated abnormal FAO and glycolysis in mice and tubular cells. CONCLUSION Our study provides experimental evidence for the use of Dapa as a means to reduce diabetic AKI by ameliorating metabolic reprogramming in renal tubular cells.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/drug therapy
- Acute Kidney Injury/pathology
- Acute Kidney Injury/etiology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/pathology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/drug therapy
- Diabetic Nephropathies/pathology
- Glucosides/pharmacology
- Glucosides/therapeutic use
- Metabolic Reprogramming/drug effects
- Mice, Inbred C57BL
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/pathology
- Signal Transduction/drug effects
- Sirtuin 3/metabolism
- Sirtuin 3/genetics
- Benzhydryl Compounds/pharmacology
- Benzhydryl Compounds/therapeutic use
Collapse
Affiliation(s)
- Huimin Li
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China; Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China; Department of Nephrology, Affiliated Renhe Hospital of China Three Gorges University, Yichang City 443001, Hubei Province, China
| | - Yao Xia
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China; Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China; Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China
| | - Yafei Zhang
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China; Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China
| | - Lang Shi
- Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China; Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - JiaYi Wang
- Department of Anesthesiology, the Second Xiangya Hospital, Changsha, Hunan Province, China
| | - Hua Huang
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China; Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China
| | - Ruchi Yue
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China; Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China; Department of Nephrology, Affiliated Renhe Hospital of China Three Gorges University, Yichang City 443001, Hubei Province, China
| | - Bin Hu
- Department of Nephrology, Affiliated Renhe Hospital of China Three Gorges University, Yichang City 443001, Hubei Province, China
| | - Jiefu Zhu
- Institute of Kidney Disease, Three Gorges University, Yichang, Hubei 443000, China; Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhixia Song
- Department of Nephrology, the Longhua District People's Hospital of Shenzhen, Shenzhen, Guangdong 518000, China.
| |
Collapse
|
37
|
Wang XL, Li L, Meng X. Interplay between the Redox System and Renal Tubular Transport. Antioxidants (Basel) 2024; 13:1156. [PMID: 39456410 PMCID: PMC11505102 DOI: 10.3390/antiox13101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The kidney plays a critical role in maintaining the homeostasis of body fluid by filtration of metabolic wastes and reabsorption of nutrients. Due to the overload, a vast of energy is required through aerobic metabolism, which inevitably leads to the generation of reactive oxygen species (ROS) in the kidney. Under unstressed conditions, ROS are counteracted by antioxidant systems and maintained at low levels, which are involved in signal transduction and physiological processes. Accumulating evidence indicates that the reduction-oxidation (redox) system interacts with renal tubular transport. Redox imbalance or dysfunction of tubular transport leads to renal disease. Here, we discuss the ROS and antioxidant systems in the kidney and outline the metabolic dysfunction that is a common feature of renal disease. Importantly, we describe the key molecules involved in renal tubular transport and their relationship to the redox system and, finally, summarize the impact of their dysregulation on the pathogenesis and progression of acute and chronic kidney disease.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Lianjian Li
- Department of Vascular Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Hubei Academy of Chinese Medicine, Wuhan 430061, China;
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
38
|
Chen Z, Chen L, Lyu TD, Weng S, Xie Y, Jin Y, Wu O, Jones M, Kwan K, Makvnadi P, Li B, Sharopov F, Ma C, Li H, Wu A. Targeted mitochondrial nanomaterials in biomedicine: Advances in therapeutic strategies and imaging modalities. Acta Biomater 2024; 186:1-29. [PMID: 39151665 DOI: 10.1016/j.actbio.2024.08.008] [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: 05/15/2024] [Revised: 07/15/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
Mitochondria, pivotal organelles crucial for energy generation, apoptosis regulation, and cellular metabolism, have spurred remarkable advancements in targeted material development. This review surveys recent breakthroughs in targeted mitochondrial nanomaterials, illuminating their potential in drug delivery, disease management, and biomedical imaging. This review approaches from various application perspectives, introducing the specific applications of mitochondria-targeted materials in cancer treatment, probes and imaging, and diseases treated with mitochondria as a therapeutic target. Addressing extant challenges and elucidating potential therapeutic mechanisms, it also outlines future development trajectories and obstacles. By comprehensively exploring the diverse applications of targeted mitochondrial nanomaterials, this review aims to catalyze innovative treatment modalities and diagnostic approaches in medical research. STATEMENT OF SIGNIFICANCE: This review presents the latest advancements in mitochondria-targeted nanomaterials for biomedical applications, covering diverse fields such as cancer therapy, bioprobes, imaging, and the treatment of various systemic diseases. The novelty and significance of this work lie in its systematic analysis of the intricate relationship between mitochondria and different diseases, as well as the ingenious design strategies employed to harness the therapeutic potential of nanomaterials. By providing crucial insights into the development of mitochondria-targeted nanomaterials and their applications, this review offers a valuable resource for researchers working on innovative treatment modalities and diagnostic approaches. The scientific impact and interest to the readership lie in the identification of promising avenues for future research and the potential for clinical translation of these cutting-edge technologies.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China
| | - Linjie Chen
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Tai Dong Lyu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Shoutao Weng
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Yihao Xie
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Yuxin Jin
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Ouqiang Wu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham B31 2AP, UK
| | - Kenny Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pooyan Makvnadi
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India; Centre of Research Impact and Outreach, Chitkara University, Rajpura, Punjab 140417, India
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College Soochow University, PR China
| | - Farukh Sharopov
- V.I. Nikitin Chemistry Institute of Tajikistan National Academy of Sciences, Dushanbe 734063, Tajikistan
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | - Huaqiong Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China.
| | - Aimin Wu
- Department of Orthopaedics Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou, Zhejiang Province 325035, PR China.
| |
Collapse
|
39
|
Guan Y, Yin X, Wang L, Diao Z, Huang H, Wang X. Biomarkers of Arginine Methylation in Diabetic Nephropathy: Novel Insights from Bioinformatics Analysis. Diabetes Metab Syndr Obes 2024; 17:3399-3418. [PMID: 39290792 PMCID: PMC11407315 DOI: 10.2147/dmso.s472412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
Background Diabetic nephropathy (DN) is a severe complication of diabetes influenced by arginine methylation. This study aimed to elucidate the role of protein arginine methylation-related genes (PRMT-RGs) in DN and identify potential biomarkers. Methods Differentially expressed genes in two GEO datasets (GSE30122 and GSE104954) were integrated with 9 PRMT-RGs. Candidate genes were identified using WGCNA and differential expression analysis, then screened using support vector machine-recursive feature elimination and least absolute shrinkage and selection operator. Biomarkers were defined as genes with consistent differential expression across both datasets. Regulatory networks were constructed using the miRNet and Network Analyst databases. Gene set enrichment analysis was performed to identify the signaling pathways in which the biomarkers were enriched in DN. Different immune cells in DN were identified using immune infiltration analysis. Meanwhile, drug prediction and molecular docking identified potential DN therapies. Finally, qRT-PCR and immunohistochemistry validated two biomarkers in STZ-induced DN mice and DN patients. Results Two biomarkers (FAM98A and FAM13B) of DN were identified in this study. The molecular regulatory network revealed that FAM98A and FAM13B were co-regulated by 6 microRNAs and 1 transcription factor and were enriched in signaling pathways. Immune infiltration and correlation analyses revealed that FAM98A and FAM13B were involved in developing DN along with PRMT-RGs and immune cells. The expression levels of Fam98a and Fam13b were significantly upregulated in the kidneys of DN mice revealed by qRT-PCR analysis. The expression levels of FAM98A were significantly upregulated in the kidneys of DN patients revealed by immunohistochemistry staining. Molecular docking showed that estradiol and rotenone exerted potential therapeutic effects on DN by targeting FAM98A. Conclusion Comprehensive bioinformatics analysis revealed that FAM98A and FAM13B were potential DN biomarkers correlated with PRMT-RGs and immune cells. This study provided useful insights for elucidating the molecular mechanisms and developing targeted therapy for DN.
Collapse
Affiliation(s)
- Yiming Guan
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiayan Yin
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Liyan Wang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hongdong Huang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xueqi Wang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
40
|
Zhang C, Shen S, Xu L, Li M, Tian B, Yao L, Zhu X. LONP1 alleviates ageing-related renal fibrosis by maintaining mitochondrial homeostasis. J Cell Mol Med 2024; 28:e70090. [PMID: 39261902 PMCID: PMC11390342 DOI: 10.1111/jcmm.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
Mitochondrial dysfunction is a pivotal event contributing to the development of ageing-related kidney disorders. Lon protease 1 (LONP1) has been reported to be responsible for ageing-related renal fibrosis; however, the underlying mechanism(s) of LONP1-driven kidney ageing with respect to mitochondrial disturbances remains to be further explored. The level of LONP1 was tested in the kidneys of aged humans and mice. Renal fibrosis and mitochondrial quality control were confirmed in the kidneys of aged mice. Effects of LONP1 silencing or overexpression on renal fibrosis and mitochondrial quality control were explored. In addition, N6-methyladenosine (m6A) modification and methyltransferase like 3 (METTL3) levels, the relationship between LONP1 and METTL3, and the impacts of METTL3 overexpression on mitochondrial functions were confirmed. Furthermore, the expression of insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) and the regulatory effects of IGF2BP2 on LONP1 were confirmed in vitro. LONP1 expression was reduced in the kidneys of aged humans and mice, accompanied by renal fibrosis and mitochondrial dysregulation. Overexpression of LONP1 alleviated renal fibrosis and maintained mitochondrial homeostasis, while silencing of LONP1 had the opposite effect. Impaired METTL3-m6A signalling contributed at least in part to ageing-induced LONP1 modification, reducing subsequent degradation in an IGF2BP2-dependent manner. Moreover, METTL3 overexpression alleviated proximal tubule cell injury, preserved mitochondrial stability, inhibited LONP1 degradation, and protected mitochondrial functions. LONP1 mediates mitochondrial function in kidney ageing and that targeting LONP1 may be a potential therapeutic strategy for improving ageing-related renal fibrosis.
Collapse
Affiliation(s)
- Congxiao Zhang
- Blood Purification CenterThe Fourth People's Hospital of Shenyang, China Medical UniversityShenyangLiaoningP. R. China
| | - Siman Shen
- Department of AnesthesiologyThe Second Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongP. R. China
| | - Li Xu
- Department of Laboratory MedicineThe Second Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongP. R. China
| | - Man Li
- Blood Purification CenterThe Fourth People's Hospital of Shenyang, China Medical UniversityShenyangLiaoningP. R. China
| | - Binyao Tian
- Department of NephrologyThe First Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Li Yao
- Department of NephrologyThe First Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Xinwang Zhu
- Department of NephrologyThe First Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| |
Collapse
|
41
|
Wang M, Li Q, Wang S, Zuo L, Hai Y, Yuan S, Li X, Huang X, Yang C, Yao L, Cao W, Zuo G, Wang J. Astragaloside IV protects renal tubular epithelial cells against oxidative stress-induced injury by upregulating CPT1A-mediated HSD17B10 lysine succinylation in diabetic kidney disease. Phytother Res 2024; 38:4519-4540. [PMID: 39038923 DOI: 10.1002/ptr.8298] [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: 12/16/2023] [Revised: 05/13/2024] [Accepted: 07/07/2024] [Indexed: 07/24/2024]
Abstract
Tubular injury and oxidative stress are involved in the pathogenesis of diabetic kidney disease (DKD). Astragaloside IV (ASIV) is a natural antioxidant. The effects and underlying molecular mechanisms of ASIV on DKD have not been elucidated. The db/db mice and high-glucose-stimulated HK2 cells were used to evaluate the beneficial effects of ASIV in vivo and in vitro. Succinylated proteomics was used to identify novel mechanisms of ASIV against DKD and experimentally further validated. ASIV alleviated renal dysfunction and proteinuria, downregulated fasting blood glucose, and upregulated insulin sensitivity in db/db mice. Meanwhile, ASIV alleviated tubular injury, oxidative stress, and mitochondrial dysfunction in vivo and in vitro. Mechanistically, ASIV reversed downregulated 17beta-hydroxysteroid dehydrogenase type 10 (HSD17B10) lysine succinylation by restoring carnitine palmitoyl-transferase1alpha (Cpt1a or CPT1A) activity in vivo and in vitro. Molecular docking and cell thermal shift assay revealed that ASIV may bind to CPT1A. Molecular dynamics simulations demonstrated K99 succinylation of HSD17B10 maintained mitochondrial RNA ribonuclease P (RNase P) stability. The K99R mutation of HSD17B10 induced oxidative stress and disrupted its binding to CPT1A or mitochondrial ribonuclease P protein 1 (MRPP1). Importantly, ASIV restored the interaction between HSD17B10 and MRPP1 in vivo and in vitro. We also demonstrated that ASIV reversed high-glucose-induced impaired RNase P activity in HK2 cells, which was suppressed upon K99R mutation of HSD17B10. These findings suggest that ASIV ameliorates oxidative stress-associated proximal tubular injury by upregulating CPT1A-mediated K99 succinylation of HSD17B10 to maintain RNase P activity.
Collapse
Affiliation(s)
- Meng Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Qiurui Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Ling Zuo
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Yang Hai
- Basic Medicine College, Chongqing Medical University, Chongqing, China
| | - Su Yuan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Xuezhi Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Xuekuan Huang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Congwen Yang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Ling Yao
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Wenfu Cao
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Guoqing Zuo
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Xiao L, Ye G. RUNX3 alleviates mitochondrial dysfunction and tubular damage by inhibiting TLR4/NF-κB signalling pathway in diabetic kidney disease. Nephrology (Carlton) 2024; 29:470-481. [PMID: 38735649 DOI: 10.1111/nep.14307] [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: 11/28/2023] [Revised: 01/16/2024] [Accepted: 04/05/2024] [Indexed: 05/14/2024]
Abstract
AIM The impaired function of tubular mitochondria is critical in diabetic kidney disease (DKD) progression. RUNX3 is down-regulated in DKD models. We intend to explore the effects of RUNX3 on mitochondrial dysfunction and renal tubule injury in DKD and related mechanisms. METHODS The development of diabetes models involved injecting mice with streptozotocin while treating HK-2 cells with high glucose (HG). By using immunohistochemical techniques, the renal localizations of RUNX3 were identified. Levels of adenosine triphosphate (ATP), mitochondrial membrane potential, and biochemical index were detected by appropriate kits. Reactive oxygen species (ROS) generation was assessed with dihydroethidium and MitoSOX Red staining. Apoptosis was assessed by flow cytometry and TUNEL. RUNX3 ubiquitination was measured. RESULTS RUNX3 was mainly present in renal tubules. Overexpressing RUNX3 increased Mfn2, Mfn1, ATP levels, and mitochondrial membrane potential, reduced Drp1 and ROS levels and cell apoptosis, as well as Cyt-C release into the cytoplasm. RUNX3 overexpression displayed a reduction in urinary albumin to creatinine ratio, Hemoglobin A1c, serum creatinine, and blood urea nitrogen. Overexpressing TLR4 attenuated the inhibitory effect of RUNX3 overexpression on mitochondrial dysfunction and cell apoptosis. HG promoted RUNX3 ubiquitination and SMURF2 expression. RUNX3 knockdown cancelled the inhibitory effect of SMURF2 on mitochondrial dysfunction and cell apoptosis. CONCLUSION SMURF2 interference inhibits RUNX3 ubiquitination and TLR4/NF-κB signalling pathway, thereby alleviating renal tubule injury.
Collapse
Affiliation(s)
- Ling Xiao
- Department of Nephrology, Wuhan Third Hospital, Wuhan, China
| | - Gang Ye
- Department of Nephrology, Wuhan Third Hospital, Wuhan, China
| |
Collapse
|
43
|
Wang D, Li Y, Li G, Liu M, Zhou Z, Wu M, Song S, Bian Y, Dong J, Li X, Du Y, Zhang T, Shi Y. Inhibition of PKC-δ retards kidney fibrosis via inhibiting cGAS-STING signaling pathway in mice. Cell Death Discov 2024; 10:314. [PMID: 38972937 PMCID: PMC11228024 DOI: 10.1038/s41420-024-02087-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Kidney fibrosis is considered to be the ultimate aggregation pathway of chronic kidney disease (CKD), but its underlying mechanism remains elusive. Protein kinase C-delta (PKC-δ) plays critical roles in the control of growth, differentiation, and apoptosis. In this study, we found that PKC-δ was highly upregulated in human biopsy samples and mouse kidneys with fibrosis. Rottlerin, a PKC-δ inhibitor, alleviated unilateral ureteral ligation (UUO)-induced kidney fibrosis, inflammation, VDAC1 expression, and cGAS-STING signaling pathway activation. Adeno-associated virus 9 (AAV9)-mediated VDAC1 silencing or VBIT-12, a VDAC1 inhibitor, attenuated renal injury, inflammation, and activation of cGAS-STING signaling pathway in UUO mouse model. Genetic and pharmacologic inhibition of STING relieved renal fibrosis and inflammation in UUO mice. In vitro, hypoxia resulted in PKC-δ phosphorylation, VDAC1 oligomerization, and activation of cGAS-STING signaling pathway in HK-2 cells. Inhibition of PKC-δ, VDAC1 or STING alleviated hypoxia-induced fibrotic and inflammatory responses in HK-2 cells, respectively. Mechanistically, PKC-δ activation induced mitochondrial membrane VDAC1 oligomerization via direct binding VDAC1, followed by the mitochondrial DNA (mtDNA) release into the cytoplasm, and subsequent activated cGAS-STING signaling pathway, which contributed to the inflammation leading to fibrosis. In conclusion, this study has indicated for the first time that PKC-δ is an important regulator in kidney fibrosis by promoting cGAS-STING signaling pathway which mediated by VDAC1. PKC-δ may be useful for treating renal fibrosis and subsequent CKD.
Collapse
Affiliation(s)
- Dongyun Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yue Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Guiying Li
- Department of Nephrology, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China
| | - Mengyu Liu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zihui Zhou
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yawei Bian
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Jiajia Dong
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Xinran Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yunxia Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Tao Zhang
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
- Department of Nephrology, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China.
| |
Collapse
|
44
|
Yang Y, Liu J, Shi Q, Guo B, Jia H, Yang Y, Fu S. Roles of Mitochondrial Dysfunction in Diabetic Kidney Disease: New Perspectives from Mechanism to Therapy. Biomolecules 2024; 14:733. [PMID: 38927136 PMCID: PMC11201432 DOI: 10.3390/biom14060733] [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: 05/11/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication of diabetes and the main cause of end-stage renal disease around the world. Mitochondria are the main organelles responsible for producing energy in cells and are closely involved in maintaining normal organ function. Studies have found that a high-sugar environment can damage glomeruli and tubules and trigger mitochondrial dysfunction. Meanwhile, animal experiments have shown that DKD symptoms are alleviated when mitochondrial damage is targeted, suggesting that mitochondrial dysfunction is inextricably linked to the development of DKD. This article describes the mechanisms of mitochondrial dysfunction and the progression and onset of DKD. The relationship between DKD and mitochondrial dysfunction is discussed. At the same time, the progress of DKD treatment targeting mitochondrial dysfunction is summarized. We hope to provide new insights into the progress and treatment of DKD.
Collapse
Affiliation(s)
- Yichen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (Y.Y.); (J.L.); (B.G.); (H.J.); (Y.Y.)
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jiahui Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (Y.Y.); (J.L.); (B.G.); (H.J.); (Y.Y.)
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Qiling Shi
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China;
| | - Buyu Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (Y.Y.); (J.L.); (B.G.); (H.J.); (Y.Y.)
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Hanbing Jia
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (Y.Y.); (J.L.); (B.G.); (H.J.); (Y.Y.)
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yuxuan Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (Y.Y.); (J.L.); (B.G.); (H.J.); (Y.Y.)
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Songbo Fu
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou 730000, China
- Gansu Provincial Endocrine Disease Clinical Medicine Research Center, Lanzhou 730000, China
| |
Collapse
|
45
|
Jia X, Zhu L, Zhu Q, Zhang J. The role of mitochondrial dysfunction in kidney injury and disease. Autoimmun Rev 2024; 23:103576. [PMID: 38909720 DOI: 10.1016/j.autrev.2024.103576] [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: 01/22/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Mitochondria are the main sites of aerobic respiration in the cell and mainly provide energy for the organism, and play key roles in adenosine triphosphate (ATP) synthesis, metabolic regulation, and cell differentiation and death. Mitochondrial dysfunction has been identified as a contributing factor to a variety of diseases. The kidney is rich in mitochondria to meet energy needs, and stable mitochondrial structure and function are essential for normal kidney function. Recently, many studies have shown a link between mitochondrial dysfunction and kidney disease, maintaining mitochondrial homeostasis has become an important target for kidney therapy. In this review, we integrate the role of mitochondrial dysfunction in different kidney diseases, and specifically elaborate the mechanism of mitochondrial reactive oxygen species (mtROS), autophagy and ferroptosis involved in the occurrence and development of kidney diseases, providing insights for improved treatment of kidney diseases.
Collapse
Affiliation(s)
- Xueqian Jia
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Lifu Zhu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Qixing Zhu
- Institute of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Key Laboratory of Dermatology, Ministry of Education, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| | - Jiaxiang Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China; Key Laboratory of Dermatology, Ministry of Education, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; The Center for Scientific Research, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
46
|
Guo X, Wang J, Wu Y, Zhu X, Xu L. Renal aging and mitochondrial quality control. Biogerontology 2024; 25:399-414. [PMID: 38349436 DOI: 10.1007/s10522-023-10091-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/29/2023] [Indexed: 06/01/2024]
Abstract
Mitochondria are dynamic organelles that participate in different cellular process that control metabolism, cell division, and survival, and the kidney is one of the most metabolically active organs that contains abundant mitochondria. Perturbations in mitochondrial homeostasis in the kidney can accelerate kidney aging, and maintaining mitochondrial homeostasis can effectively delay aging in the kidney. Kidney aging is a degenerative process linked to detrimental processes. The significance of aberrant mitochondrial homeostasis in renal aging has received increasing attention. However, the contribution of mitochondrial quality control (MQC) to renal aging has not been reviewed in detail. Here, we generalize the current factors contributing to renal aging, review the alterations in MQC during renal injury and aging, and analyze the relationship between mitochondria and intrinsic renal cells. We also introduce MQC in the context of renal aging, and discuss the study of mitochondria in the intrinsic cells of the kidney, which is the innovation of our paper. In addition, during kidney injury and repair, the specific functions and regulatory mechanisms of MQC systems in resident and circulating cell types remain unclear. Currently, most of the studies we reviewed are based on animal and cellular models, the relationship between renal tissue aging and mitochondria has not been adequately investigated in clinical studies, and there is still a long way to go.
Collapse
Affiliation(s)
- Xiuli Guo
- Department of Laboratory, The First Hospital of China Medical University, Shenyang, China
| | - Jiao Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yinjie Wu
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Xinwang Zhu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Li Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524003, Guangdong, People's Republic of China.
| |
Collapse
|
47
|
Xie T, Yao L, Li X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants (Basel) 2024; 13:659. [PMID: 38929098 PMCID: PMC11200795 DOI: 10.3390/antiox13060659] [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: 05/06/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney diseases pose a significant global health issue, frequently resulting in the gradual decline of renal function and eventually leading to end-stage renal failure. Abnormal iron metabolism and oxidative stress-mediated cellular dysfunction facilitates the advancement of kidney diseases. Iron homeostasis is strictly regulated in the body, and disturbance in this regulatory system results in abnormal iron accumulation or deficiency, both of which are associated with the pathogenesis of kidney diseases. Iron overload promotes the production of reactive oxygen species (ROS) through the Fenton reaction, resulting in oxidative damage to cellular molecules and impaired cellular function. Increased oxidative stress can also influence iron metabolism through upregulation of iron regulatory proteins and altering the expression and activity of key iron transport and storage proteins. This creates a harmful cycle in which abnormal iron metabolism and oxidative stress perpetuate each other, ultimately contributing to the advancement of kidney diseases. The crosstalk of iron metabolism and oxidative stress involves multiple signaling pathways, such as hypoxia-inducible factor (HIF) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. This review delves into the functions and mechanisms of iron metabolism and oxidative stress, along with the intricate relationship between these two factors in the context of kidney diseases. Understanding the underlying mechanisms should help to identify potential therapeutic targets and develop novel and effective therapeutic strategies to combat the burden of kidney diseases.
Collapse
Affiliation(s)
- Tiancheng Xie
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
48
|
Guo Y, Che R, Wang P, Zhang A. Mitochondrial dysfunction in the pathophysiology of renal diseases. Am J Physiol Renal Physiol 2024; 326:F768-F779. [PMID: 38450435 DOI: 10.1152/ajprenal.00189.2023] [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: 07/12/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/08/2024] Open
Abstract
Mitochondria are essential organelles in the human body, serving as the metabolic factory of the whole organism. When mitochondria are dysfunctional, it can affect all organs of the body. The kidney is rich in mitochondria, and its function is closely related to the development of kidney diseases. Studying the relationship between mitochondria and kidney disease progression is of great interest. In the past decade, scientists have made inspiring progress in investigating the role of mitochondria in the pathophysiology of renal diseases. This article discusses various mechanisms for maintaining mitochondrial quality, including mitochondrial energetics, mitochondrial biogenesis, mitochondrial dynamics, mitochondrial DNA repair, mitochondrial proteolysis and the unfolded protein response, mitochondrial autophagy, mitochondria-derived vesicles, and mitocytosis. The article also highlights the cross talk between mitochondria and other organelles, with a focus on kidney diseases. Finally, the article concludes with an overview of mitochondria-related clinical research.
Collapse
Affiliation(s)
- Yuxian Guo
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Ruochen Che
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Peipei Wang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
49
|
Huo Z, Gu J, He T. Apelin‑13 reduces high glucose‑induced mitochondrial dysfunction in cochlear hair cells by inhibiting endoplasmic reticulum stress. Exp Ther Med 2024; 27:226. [PMID: 38596659 PMCID: PMC11002831 DOI: 10.3892/etm.2024.12515] [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: 10/31/2023] [Accepted: 02/28/2024] [Indexed: 04/11/2024] Open
Abstract
The complex manifestation of diabetic hearing loss and the relative inaccessibility of the inner ear contribute to the lack of research. The present study aimed to reveal the role of Apelin-13, a critical regulator of lipid metabolism, in diabetes-induced hearing loss. Cochlear hair cells treated with high glucose (HG) were adopted as an in vitro research model, and the impacts of Apelin-13 on cellular oxidative stress, apoptosis, mitochondrial dysfunction and endoplasmic reticulum (ER) stress were determined. In addition, cells were treated with the ER stress agonist tunicamycin to further explore its potential role in the regulatory effects of Apelin-13. Apelin-13 inhibited oxidative stress and apoptosis in the HG-induced cells. Additionally, Apelin-13 elevated mitochondrial membrane potential and ATP production, whereas it reduced mitochondrial reactive oxygen species levels. The levels of ER stress-related proteins exhibited a downward trend in response to Apelin-13. By contrast, tunicamycin reversed the effects of Apelin-13 on the aforementioned aspects, suggesting the role of ER stress in the regulatory effects of Apelin-13. In conclusion, the present study elucidated the protective role of Apelin-13 in ameliorating HG-induced mitochondrial functional impairment in cochlear hair cells by inhibiting ER stress.
Collapse
Affiliation(s)
- Zhiqiang Huo
- Department of Otolaryngology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| | - Jun Gu
- Department of Otolaryngology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| | - Teng He
- Department of Otolaryngology, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu 215500, P.R. China
| |
Collapse
|
50
|
Athinarayanan SJ, Roberts CGP, Vangala C, Shetty GK, McKenzie AL, Weimbs T, Volek JS. The case for a ketogenic diet in the management of kidney disease. BMJ Open Diabetes Res Care 2024; 12:e004101. [PMID: 38677719 PMCID: PMC11057262 DOI: 10.1136/bmjdrc-2024-004101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
Ketogenic diets have been widely used for weight loss and are increasingly used in the management of type 2 diabetes. Despite evidence that ketones have multiple positive effects on kidney function, common misconceptions about ketogenic diets, such as high protein content and acid load, have prevented their widespread use in individuals with impaired kidney function. Clinical trial evidence focusing on major adverse kidney events is sparse. The aim of this review is to explore the effects of a ketogenic diet, with an emphasis on the pleiotropic actions of ketones, on kidney health. Given the minimal concerns in relation to the potential renoprotective effects of a ketogenic diet, future studies should evaluate the safety and efficacy of ketogenic interventions in kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas Weimbs
- Department of Molecular Cellular & Developmental Biology, University of California Santa Barbara, Santa Barbara, California, USA
| | - Jeff S Volek
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|